US20030147907A1 - Retroviral vectors - Google Patents

Retroviral vectors Download PDF

Info

Publication number
US20030147907A1
US20030147907A1 US09/867,947 US86794701A US2003147907A1 US 20030147907 A1 US20030147907 A1 US 20030147907A1 US 86794701 A US86794701 A US 86794701A US 2003147907 A1 US2003147907 A1 US 2003147907A1
Authority
US
United States
Prior art keywords
vector
retroviral vector
eiav
retroviral
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US09/867,947
Inventor
Alan Kingsman
Miles Carroll
Jonathan Rohll
Kyriacos Mitrophanous
Narry Kim
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
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
Priority claimed from GBGB9727135.7A external-priority patent/GB9727135D0/en
Priority claimed from GBGB9811037.2A external-priority patent/GB9811037D0/en
Application filed by Individual filed Critical Individual
Priority to US09/867,947 priority Critical patent/US20030147907A1/en
Publication of US20030147907A1 publication Critical patent/US20030147907A1/en
Priority to US11/154,421 priority patent/US20060024274A1/en
Priority to US11/583,427 priority patent/US20070213290A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24141Use of virus, viral particle or viral elements as a vector
    • C12N2710/24143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic 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
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24141Use of virus, viral particle or viral elements as a vector
    • C12N2710/24144Chimeric viral vector comprising heterologous viral elements for production of another viral 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15051Methods of production or purification of viral material
    • C12N2740/15052Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15061Methods of inactivation or attenuation
    • C12N2740/15062Methods of inactivation or attenuation by genetic engineering
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/60Vectors comprising as targeting moiety peptide derived from defined protein from viruses
    • C12N2810/6072Vectors comprising as targeting moiety peptide derived from defined protein from viruses negative strand RNA viruses
    • C12N2810/6081Vectors comprising as targeting moiety peptide derived from defined protein from viruses negative strand RNA viruses rhabdoviridae, e.g. VSV

Definitions

  • This invention relates to a retroviral vector.
  • it relates to retroviral vectors capable of transferring genetic material to non-dividing or slowly-dividing cells derived from non-primate lentiviruses.
  • lentiviral vectors enable very stable long-term expression of the gene of interest. This has been shown to be at least three months for transduced rat neuronal cells.
  • the MLV based vectors were only able to express the gene of interest for six weeks.
  • HIV-based vectors produced to date result in an integrated provirus in the transduced cell that has HIV LTRs at its ends. This limits the use of these vectors as the LTRs have to be used as expression signals for any inserted gene unless an internal promoter is used.
  • the use of internal promoters has significant disadvantages. The unpredictable outcome of placing additional promoters within the retroviral LTR transcription unit is well documented (Bowtell et al, 1988 J. Virol. 62, 2464; Correll et al, 1994 Blood 84, 1812; Emerman and Temin 1984 Cell 39, 459; Ghattas et al, 1991 Mol. Cell. Biol.
  • the factors involved appear to include the relative position and orientation of the two promoters, the nature of the promoters and the expressed genes and any selection procedures that may be adopted.
  • the presence of internal promoters can affect both the transduction titers attainable from a packaging cell line and the stability of the integrated vector.
  • HIV and other lentiviral LTRs have virus-specific requirements for gene expression.
  • the HIV LTR is not active in the absence of the viral Tat protein (Cullen 1995 AIDS 9, S19). It is desirable, therefore, to modify the LTRs in such a way as to change the requirements for gene expression. In particular tissue specific gene expression signals may be required for some gene therapy applications.
  • HIV vectors have a number of significant disadvantages which may limit their therapeutic application to certain diseases. HIV-1 has the disadvantage of being a human pathogen carrying potentially oncogenic proteins and sequences. There is the risk that introduction of vector particles produced in packaging cells which express HIV gag-pol will introduce these proteins into the patient leading to seroconversion. For these reasons, there is a need to develop lentiviral-based vectors which do not introduce HIV proteins into patients.
  • Retroviruses are limited in the length of RNA sequences which can be packaged efficiently and so the existence of long regions of the retroviral genome severely limits the coding capacity of the vector for heterologous coding RNA.
  • the sequence requirements for packaging HIV vector genomes are complex.
  • the HIV-1 packaging signal encompasses the splice donor site and contains a portion of the 5′-untranslated region of the gag gene which has a putative secondary structure containing 4 short stem-loops. Additional sequences elsewhere in the genome are also known to be important for efficient encapsidation of HV. For example the first 350 bps of the gag protein coding sequence may contribute to efficient packaging and ill defined regions of env are also implicated.
  • a packaging signal extending to 350 bps of the gag protein-coding region has been used.
  • a retroviral vector genome containing a deleted gag gene from a non-primate lentivirus wherein the deletion in gag removes one or more nucleotides downstream of nucleotide 350 of the gag coding sequence.
  • the deletion extends from nucleotide 350 to at least the C-terminus of the gagpol coding region. More preferably the deletion additionally removes nucleotide 300 of the gag coding region and most preferably the deletion retains only the first 150 nucleotides of the gag coding region.
  • the gag coding region contains the first 109 nucleotides of the gag coding region. It may also be possible for the gag coding region to contain only the first 2 nucleotides of the gag coding region.
  • the lentiviral genome may contain accessory genes derived from a retrovirus, such as, but not limited to, a rev gene, a tat gene, a vif gene, a nef gene, a vpr gene or an S2 gene. Additional components may be added such as introns, splice-donor sites, a rev responsive element (RRE), cloning sites and selectable marker genes.
  • RRE rev responsive element
  • non-primate lentivirus genome from which the vector is derived lacks one or more accessory genes.
  • accessory genes are highly advantageous. Firstly, it permits vectors to be produced without the genes normally associated with disease in lentiviral (e.g. HIV) infections. In particular, tat and env are associated with disease. Secondly, the deletion of accessory genes permits the vector to package more heterologous DNA. Thirdly, genes whose function is unknown, such as dUTPase and S2, may be omitted, thus reducing the risk of causing undesired effects.
  • leader sequence of the non-primate lentivirus genome is essential for high protein expression of gag and gagpol.
  • non-primate lentivirus genome from which the vector is derived lacks the tat gene but includes the leader sequence between the end of the 5′ LTR and the ATG of gag.
  • a vector is provided with maximal genetic capacity and high titre, but without accessory genes that are either of unknown function (S2, UTPase), and therefore may present risk, or are analogues of HIV proteins that may be associated with AIDS (tat, env).
  • the present invention provides a retroviral vector derived from a non-primate lentivirus genome (1) comprising a deleted gag gene wherein the deletion in gag removes one or more nucleotides downstream of nucleotide 350 of the gag coding sequence; (2) wherein one or more accessory genes are absent from the non-primate lentivirus genome; (3) wherein the non-primate lentivirus genome lacks the tat gene but includes the leader sequence between the end of the 5 ⁇ LTR and the ATG of gag; and combinations of (1), (2) and (3).
  • the retroviral vector comprises all of features (1) and (2) and (3).
  • non-primate vector refers to a vector derived from a virus which does not primarily infect primates, especially humans.
  • non-primate virus vectors include vectors which infect non-primate mammals, such as dogs, sheep and horses, reptiles, birds and insects.
  • a lentiviral or lentivirus vector is a vector which comprises at least one component part derived from a lentivirus. Preferably, that component part is involved in the biological mechanisms by which the vector infects cells, expresses genes or is replicated.
  • the non-primate lentivirus may be any member of the family of lentiviridae which does not naturally infect a primate and may include a feline immunodeficiency virus (F1v), a bovine immunodeficiency virus (BIV), a caprine arthritis encephalitis virus (CAEV), a Maedi visna virus (MVV) or an equine infectious anaemia virus (EIAV).
  • F1v feline immunodeficiency virus
  • BIV bovine immunodeficiency virus
  • CAEV caprine arthritis encephalitis virus
  • MVV Maedi visna virus
  • EIAV equine infectious anaemia virus
  • the lentivirus is an EIAV.
  • Equine infectious anaemia virus infects all equidae resulting in plasma viremia and thrombocytopenia (Clabough, et al. 1991. J Virol. 65:6242-51). Virus replication is thought to be controlled by the process of matur
  • EIAV has the simplest genomic structure of the lentiviruses.
  • EIAV encodes three other genes: tat, rev, and S2.
  • Tat acts as a transcriptional activator of the viral LTR (Derse and Newboldl993 Virology. 194:530-6; Maury, et al 1994 Virology. 200:632-42.) and Rev regulates and coordinates the expression of viral genes through rev-response elements (RRE) (Martarano et al 1994 J Virol. 68:3102-11.).
  • RRE rev-response elements
  • the mechanisms of action of these two proteins are thought to be broadly similar to the analogous mechanisms in the primate viruses (Martano et al ibid).
  • S2 is unknown.
  • Ttm an EIAV protein, Ttm, has been identified that is encoded by the first exon of tat spliced to the env coding sequence at the start of the transmembrane protein.
  • reverse transcriptase and integrase non-primate lentiviruses contain a fourth pol gene product which codes for a dUTPase. This may play a role in the ability of these lentiviruses to infect certain non-dividing cell types.
  • the viral RNA in the first aspect of the invention is transcribed from a promoter, which may be of viral or non-viral origin, but which is capable of directing expression in a eukaryotic cell such as a mammalian cell.
  • a promoter which may be of viral or non-viral origin, but which is capable of directing expression in a eukaryotic cell such as a mammalian cell.
  • an enhancer is added, either upstream of the promoter or downstream.
  • the RNA transcript is terminated at a polyadenylation site which may be the one provided in the lentiviral 3 ⁇ LTR or a different polyadenylation signal.
  • the present invention provides a DNA transcription unit comprising a promoter and optionally an enhancer capable of directing expression of a retroviral vector genome.
  • Transcription units as described herein comprise regions of nucleic acid containing sequences capable of being transcribed.
  • sequences encoding MRNA, tRNA and rRNA are included within this definition.
  • the sequences may be in the sense or antisense orientation with respect to the promoter.
  • Antisense constructs can be used to inhibit the expression of a gene in a cell according to well-known techniques.
  • Nucleic acids may be, for example, ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or analogues thereof. Sequences encoding mRNA will optionally include some or all of 5′ and/or 3′ transcribed but untranslated flanking sequences naturally, or otherwise, associated with the translated coding sequence. It may optionally further include the associated transcriptional control sequences normally associated with the transcribed sequences, for example transcriptional stop signals, polyadenylation sites and downstream enhancer elements. Nucleic acids may comprise cDNA or genomic DNA (which may contain introns).
  • promoter is used in the normal sense of the art, e.g. an RNA polymerase binding site in the Jacob-Monod theory of gene expression.
  • the term “enhancer” includes a DNA sequence which binds to other protein components of the transcription initiation complex and thus facilitates the initiation of transcription directed by its associated promoter.
  • the promoter and enhancer of the transcription units encoding the first viral vector component are preferably strongly active, or capable of being strongly induced, in the producer cell under conditions for production of the retroviral vector of the present invention and/or in primary target cells under conditions for production of the secondary viral vector.
  • the promoter and enhancer of the transcription units encoding the second viral vector component are preferably strongly active, or capable of being strongly induced, in the target cells.
  • the promoter and/or enhancer may be constitutively efficient, or may be tissue or temporally restricted in their activity. Examples of suitable tissue restricted promoters/enhancers are those which are highly active in tumour cells such as a promoter/enhancer from a MUC1 gene, a CEA gene or a 5T4 antigen gene.
  • temporally restricted promoters/enhancers are those which are responsive to ischaemia and/or hypoxia, such as hypoxia response elements or the promoter/enhancer of a grp78 or a grp94 gene.
  • One preferred promoter-enhancer combination is a human cytomegalovirus (hCMV) major immediate early (MIE) promoter/enhancer combination.
  • the LTRs may be altered in, for example, U3 (such as to obtain strong constitutive expression, inducible expression or tissue specific expression); R (such as to remove TAR stem loops); or U5 (such as to use enhanced non-U5 based polyadenylation signals, for example from the bovine growth hormone gene).
  • U3 such as to obtain strong constitutive expression, inducible expression or tissue specific expression
  • R such as to remove TAR stem loops
  • U5 such as to use enhanced non-U5 based polyadenylation signals, for example from the bovine growth hormone gene.
  • the internal promoter cassette is reversed and a polyadenylation signal is placed downstream of the cassette.
  • the polyadenylation signal which is used contains at least one intron.
  • the vector of the present invention may make use of self-inactivating strategies.
  • Self-inactivating retroviral vectors have been constructed by deleting the transcriptional enhancers or the enhancers and promoters in the U3 region of the 3′ LTR. After one round of vector replication, these changes are copied into both the 5′ and the 3′ LTRs producing an inactive provirus. However, any promoters internal to the LTRs in such vectors will still be active.
  • This strategy has been employed to eliminate effects of the enhancers and promoters in the viral LTRs on transcription from internally placed genes. Such effects include increased transcription or suppression of transcription.
  • This strategy can also be used to eliminate downstream transcription from the 3′ LTR into genomic DNA. This is of particular concern in human gene therapy where it is of critical importance to prevent any activation of an endogenous oncogene.
  • Another type of self-inactivating vector has been constructed that has direct repeats flanking the packaging signal such that the packaging signal is frequently deleted during reverse transcription, producing virus defective for packaging. With sufficiently long direct repeats, a majority of resultant proviruses lose their packaging sequences. The rate of deletion could be increased to 100% by designing the vector so that packaging signal deletion reconstituted the neo marker nad be selecting the vector-infected cells in G418. This strategy may be particularly useful for gene therapy applications where any spread of the vector following gene transfer is undesirable.
  • one or more nucleotides of interest is introduced into the vector at the cloning site.
  • NOI nucleotides of interest
  • Such therapeutic genes may be expressed from a promoter placed in the retroviral LTR or may be expressed from an internal promoter introduced at the cloning site.
  • Suitable NOI coding sequences include those that are of therapeutic and/or diagnostic application such as, but are not limited to: sequences encoding cytokines, chemokines, hormones, antibodies, engineered immunoglobulin-like molecules, a single chain antibody, fusion proteins, enzymes, immune co-stimulatory molecules, immunomodulatory molecules, anti-sense RNA, a transdominant negative mutant of a target protein, a toxin, a conditional toxin, an antigen, a tumour suppressor protein and growth factors, membrane proteins, vasoactive proteins and peptides, anti-viral proteins and ribozymes, and derivatives therof (such as with an associated reporter group).
  • a suitable promoter which may be a promoter driving expression of a ribozyme(s), or a different promoter or promoters.
  • the NOI coding sequence may encode a fusion protein or a segment of a coding sequence.
  • the retroviral vector of the present invention may be used to deliver a NOI such as a pro-drug activating enzyme to a tumour site for the treatment of a cancer.
  • a suitable pro-drug is used in the treatment of the individual (such as a patient) in combination with the appropriate pro-drug activating enzyme.
  • An appropriate pro-drug is administered in conjunction with the vector.
  • pro-drugs examples include: etoposide phosphate (with alkaline phosphatase, Senter et al 1988 Proc Natl Acad Sci 85: 4842-4846); 5-fluorocytosine (with cytosine deaminase, Mullen et al 1994 Cancer Res 54: 1503-1506); Doxorubicin-N-p-hydroxyphenoxyacetamide (with Penicillin-V-Amidase, Kerr et al 1990 Cancer Immunol Immunother 31: 202-206); Para-N-bis(2-chloroethyl) aminobenzoyl glutamate (with carboxypeptidase G2); Cephalosporin nitrogen mustard carbamates (with ⁇ -lactamase); SR4233 (with P450 Reducase); Ganciclovir (with HSV thymidine kinase, Borrelli et al 1988 Proc Natl Acad Sci 85: 7572-7576); mustard pro-drugs with nitroreducta
  • the vector of the present invention may be delivered to a target site by a viral or a non-viral vector.
  • a vector is a tool that allows or faciliates the transfer of an entity from one environment to another.
  • some vectors used in recombinant DNA techniques allow entities, such as a segment of DNA (such as a heterologous DNA segment, such as a heterologous cDNA segment), to be transferred into a target cell.
  • the vector may then serve to maintain the heterologous DNA within the cell or may act as a unit of DNA replication.
  • examples of vectors used in recombinant DNA techniques include plasmids, chromosomes, artificial chromosomes or viruses.
  • Non-viral delivery systems include but are not limted to DNA transfection methods.
  • transfection includes a process using a non-viral vector to deliver a gene to a target mammalian cell.
  • Typical transfection methods include electroporation, DNA biolistics, lipid-mediated transfection, compacted DNA-mediated transfection, liposomes, immunoliposomes, lipofectin, cationic agent-mediated, cationic facial amphiphiles (CFAs) (Nature Biotechnology 1996 14; 556), and combinations thereof.
  • CFAs cationic facial amphiphiles
  • Viral delivery systems include but are not limited to adenovirus vector, an adeno-associated viral (AAV) vector, a herpes viral vector, retroviral vector, lentiviral vector, baculoviral vector.
  • Other examples of vectors include ex vivo delivery systems, which include but are not limited to DNA transfection methods such as electroporation, DNA biolistics, lipid-mediated transfection, compacted DNA-mediated transfection.
  • retroviral vector particle refers to the packaged retroviral vector, that is preferably capable of binding to and entering target cells.
  • the components of the particle may be modified with respect to the wild type retrovirus.
  • the Env proteins in the proteinaceous coat of the particle may be genetically modified in order to alter their targeting specificity or achieve some other desired function.
  • the viral vector preferentially transduces a certain cell type or cell types.
  • the viral vector is a targeted vector, that is it has a tissue tropism which is altered compared to the native virus, so that the vector is targeted to particular cells.
  • the Env protein of the retroviral secondary vector needs to be a non-toxic envelope or an envelope which may be produced in non-toxic amounts within the primary target cell, such as for example a MMLV amphotropic envelope or a modified amphotropic envelope.
  • the safety feature in such a case is preferably the deletion of regions or sequence homology between retroviral components.
  • the envelope is one which allows transduction of human cells.
  • suitable env genes include, but are not limited to, VSV-G, a MLV amphotropic env such as the 4070A env, the-RD114 feline leukaemia virus env or haemagglutinin (HA) from an influenza virus.
  • the Env protein may be one which is capable of binding to a receptor on a limited number of human cell types and may be an engineered envelope containing targeting moieties.
  • the env and gag-pol coding sequences are transcribed from a promoter and optionally an enhancer active in the chosen packaging cell line and the transcription unit is terminated by a polyadenylation signal.
  • a suitable promoter-enhancer combination is that from the human cytomegalovirus major immediate early (hCMV-MIE) gene and a polyadenylation signal from SV40 virus may be used.
  • hCMV-MIE human cytomegalovirus major immediate early
  • polyadenylation signal from SV40 virus
  • the packaging cell may be an in vivo packaging cell in the body of an individual to be treated-orit may be a cell cultured in vitro such as a tissue culture cell line. Suitable cell lines include mammalian cells such as murine fibroblast derived cell lines or human cell lines. Preferably the packaging cell line is a human cell line, such as for example: 293 cell line, HEK293, 293-T, TE671, HT1080.
  • the packaging cell may be a cell derived from the individual to be treated such as a monocyte, macrophage, stem cells, blood cell or fibroblast.
  • the cell may be isolated from an individual and the packaging and vector components administered ex vivo followed by re-administration of the autologous packaging cells.
  • the packaging and vector components may be administered to the packaging cell in vivo.
  • Methods for introducing retroviral packaging and vector components into cells of an individual are known in the art. For example, one approach is to introduce the different DNA sequences that are required to produce a retroviral vector particle e.g.
  • the vector configurations of the present invention use as their production system, three transcription units expressing a genome, the gag-pol components and an envelope.
  • the envelope expression cassette may include one of a number of envelopes such as VSV-G or various murine retrovirus envelopes such as 4070A.
  • these three cassettes would be expressed from three plasmids transiently transfected into an appropriate cell line such as 293T or from integrated copies in a stable producer cell line.
  • An alternative approach is to use another virus as an expression system for the three cassettes, for example baculovirus or adenovirus. These are both nuclear expression systems.
  • baculovirus or adenovirus are both nuclear expression systems.
  • a poxvirus to express all of the components of a retroviral or lentiviral vector system has not been described.
  • RNA handling systems such as the rev/RRE system it has not been clear whether incorporation of all three cassettes and their subsequent expression in a vector that expresses in the cytoplasm rather than the nucleus is feasible.
  • a hybrid viral vector system for in vivo gene delivery which system comprises a primary viral vector which is obtainable from or is based on a poxvirus and a second viral vector which is obtainable from or is based on a vectroviral vector, preferably a lentiviral vector, even more preferably a non-primate lentiviral vector.
  • the secondary vector may be produced from expression of essential genes for retroviral vector production encoded in the DNA of the primary vector.
  • genes may include a gag-pol from a retrovirus, an env gene from an enveloped virus and a defective retroviral vector containing one or more therapeutic or diagnostic NOI(s).
  • the defective retroviral vector contains in general terms sequences to enable reverse transcription, at least part of a 5′ long terminal repeat (LTR), at least part of a 3′LTR and a packaging signal.
  • LTR 5′ long terminal repeat
  • that secondary vector may be encoded by a plurality of transcription units, which may be located in a single or in two or more adenoviral or other primary vectors.
  • MVA-EIAV hybrids are delivered directly into the patient/animal e.g. MVA-EIAV is injected intravenously into the tail vein of a mouse and this recombinant virus infects a variety of murine tissues e.g. lung, spleen etc.
  • Infected cells express transduction competent EIAV containing a therapeutic gene for gene therapy for example.
  • EIAV vector particles bud from these cells and transduce neighbouring cells. The transduced cell then contains an integrated copy of the EIAV vector genome and expresses the therapeutic gene product or other gene product of interest.
  • a specific promoter e.g. the hypoxic response element (HRE)
  • HRE hypoxic response element
  • MVA-EIAV may be given as an aerosol delivered intranasally.
  • macrophages can be transduced in vitro and then reintroduced to create macrophage factories for EIAV-based vectors.
  • MVA is replication incompetent MVA-EIAV hybrids could also be used to treat immuno-suppressed hosts.
  • Vaccinia virus the prototypic member of the orthopox genus within the family poxviridae, was the first virus used for expression of recombinant exogenous proteins (Mackett et al 1982, Paoletti & Panicalli 1982). Vaccinia virus has a large DNA genome of greater than 180 kb and reports indicate that it can accommodate over 25 kb of foreign DNA (Merchlinsky & Moss 1992). Several other strains of poxviruses have been adapted as recombinant expression vectors (for review see Carroll and Moss 1997) e.g.
  • MVA was derived from a replication competent vaccinia smallpox vaccine strain, Ankara. After >500 passages in chick embryo fibroblast cells the virus isolate was shown to be highly attenuated in a number of animal models including mice that were immune deficient (Mayr et al 1978). The attenuated isolate, MVA, was used to vaccinate over 120,000 people, many of which were immunocompromised (Mahnel 1994) without adverse effects. Studies illustrate that MVA can infect a wide range of mammalian cells but productive infection has only been observed in Hamster kidney cell BHK-21 (Carroll 1997).
  • Retrovirus vector systems e.g. MLV-HIV and lentivirus vector systems requires the construction of producer lines that express the virus genome and essential structural proteins to make transduction competent virus. Generally, this is a relatively inefficient process which is further complicated when the virus is pseudotyped with toxic envelope proteins such as VSV-G. Expression of a functional genome and the required structural proteins from within a recombinant poxvirus may obviate many of the current inefficient retrovirus and lentivirus vector production technologies. Additionally, such recombinant poxviruses may be directly injected into patients to give rise to in vivo production of retrovirus or lentivirus.
  • MVA is a particularly suitable poxvirus for the construction of a pox-retrovirus or pox-lentivirus hybrid due to its non-replicating phenotype and its ability to perform both early and strong late expression for the production of high titre vector preparations.
  • RNA genome In order to produce a functional retrovirus or lentivirus vector genome it is essential that the 5′ of the RNA genome should be exact (Cannon et al. 1996). This is a challenge in a vaccinia-based production system as many of the vaccinia promoter comprise downstream determinants of transcription efficiency (Davison 1989b, Moss 1996). However, we show that thereare several ways to solve this problem:
  • T7 dependent system The advantage of having a T7 dependent system is that it would require the infection of the cell by two recombinant vaccinia viruses to produce transducing EIAV viral particles.
  • one MVA could carry the vector genome, under the control of the T7 promoter and the gag/pol and the env sequences under the control of the vaccinia promoters.
  • the other MVA would carry the T7 polymerase gene under the control of a vaccinia promoter (Wyatt et al 1995).
  • the retroviral vector particle according to the invention will also be capable of transducing cells which are slowly-dividing, and which non-lentiviruses such as MLV would not be able to efficiently transduce.
  • Slowly-dividing cells divide once in about every three to four days including certain tumour cells.
  • tumours contain rapidly dividing cells, some tumour cells especially those in the centre of the tumour, divide infrequently.
  • the target cell may be a growth-arrested cell capable of undergoing cell division such as a cell in a central portion of a tumour mass or a stem cell such as a haematopoietic stem cell or a CD34-positive cell.
  • the target cell may be a precursor of a differentiated cell such as a monocyte precursor, a CD33-positive cell, or a myeloid precursor.
  • the target cell may be a differentiated cell such as a neuron, astrocyte, glial cell, microglial cell, macrophage, monocyte, epithelial cell, endothelial cell or hepatocyte.
  • Target cells may be transduced either in vitro after isolation from a human individual or may be transduced directly in vivo.
  • the delivery of one or more therapeutic genes by a vector system according to the present invention may be used alone or in combination with other treatments or components of the treatment.
  • the retroviral vector of the present invention may be used to deliver one or more NOI(s) useful in the treatment of the disorders listed in WO-A-98/05635.
  • cancer inflammation or inflammatory disease
  • dermatological disorders fever, cardiovascular effects, haemorrhage, coagulation and acute phase response, cachexia, anorexia, acute infection, HIV infection, shock states, graft-versus-host reactions, autoimmune disease, reperfusion injury, meningitis, migraine and aspirin-dependent anti-thrombosis
  • cerebral ischaemia ischaemic heart disease, osteoarthritis, rheumatoid arthritis, osteoporosis, asthma, multiple sclerosis, neurodegeneration, Alzheimer's disease, atherosclerosis, stroke, vasculitis, Crohn's disease and ulcerative colitis
  • periodontitis gingiditis
  • the retroviral vector of the present invention may be used to deliver one or more NOI(s) useful in the treatment of disorders listed in WO-A-98/07859.
  • cytokine and cell proliferation/differentiation activity e.g. for treating immune deficiency, including infection with human immune deficiency virus; regulation of lymphocyte growth; treating cancer and many autoimmune diseases, and to prevent transplant rejection or induce tumour immunity
  • haematopoiesis e.g. treatment of myeloid or lymphoid diseases
  • promoting growth of bone, cartilage, tendon, ligament and nerve tissue e.g.
  • follicle-stimulating hormone for healing wounds, treatment of burns, ulcers and periodontal disease and neurodegeneration; inhibition or activation of follicle-stimulating hormone (modulation of fertility); chemotactic/chemokinetic activity (e.g. for mobilising specific cell types to sites of injury or infection); haemostatic and thrombolytic activity (e.g. for treating haemophilia and stroke); antiinflammatory activity (for treating e.g. septic shock or Crohn's disease); as antimicrobials; modulators of e.g. metabolism or behaviour; as analgesics; treating specific deficiency disorders; in treatment of e.g. psoriasis, in human or veterinary medicine.
  • the retroviral vector of the present invention may be used to deliver one or more NOI(s) useful in the treatment of disorders listed in WO-A-98/09985.
  • NOI(s) useful in the treatment of disorders listed in WO-A-98/09985.
  • macrophage inhibitory and/or T cell inhibitory activity and thus, anti-inflammatory activity i.e.
  • inhibitory effects against a cellular and/or humoral immune response including a response not associated with inflammation; inhibit the ability of macrophages and T cells to adhere to extracellular matrix components and fibronectin, as well as up-regulated fas receptor expression in T cells; inhibit unwanted immune reaction and inflammation including arthritis, including rheumatoid arthritis, inflammation associated with hypersensitivity, allergic reactions, asthma, systemic lupus erythematosus, collagen diseases and other autoimmune diseases, inflammation associated with atherosclerosis, arteriosclerosis, atherosclerotic heart disease, reperfusion injury, cardiac arrest, myocardial infarction, vascular inflammatory disorders, respiratory distress syndrome or other cardiopulmonary diseases, inflammation associated with peptic ulcer, ulcerative colitis and other diseases of the gastrointestinal tract, hepatic fibrosis, liver cirrhosis or other hepatic diseases, thyroiditis or other glandular diseases, glomerulonephritis or other renal and urologic diseases, otitis or other oto-rhino-
  • retinitis or cystoid macular oedema retinitis or cystoid macular oedema, sympathetic ophthalmia, scleritis, retinitis pigmentosa, immune and inflammatory components of degenerative fondus disease, inflammatory components of ocular trauma, ocular inflammation caused by infection, proliferative vitreo-retinopathies, acute ischaemic optic neuropathy, excessive scarring, e.g.
  • monocyte or leukocyte proliferative diseases e.g. leukaemia
  • monocytes or lymphocytes for the prevention and/or treatment of graft rejection in cases of transplantation of natural or artificial cells, tissue and organs such as cornea, bone marrow, organs, lenses, pacemakers, natural or artificial skin tissue.
  • the present invention also provides a pharmaceutical composition for treating an individual by gene therapy, wherein the composition comprises a therapeutically effective amount of the retroviral vector of the present invention comprising one or more deliverable therapeutic and/or diagnostic NOI(s) or a viral particle produced by or obtained from same.
  • the pharmaceutical composition may be for human or animal usage. Typically, a physician will determine the actual dosage which will be most suitable for an individual subject and it will vary with the age, weight and response of the particular individual.
  • the composition may optionally comprise a pharmaceutically acceptable carrier, diluent, excipient or adjuvant.
  • a pharmaceutically acceptable carrier diluent, excipient or adjuvant.
  • the choice of pharmaceutical carrier, excipient or diluent can be selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the pharmaceutical compositions may comprise as—or in addition to—the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s), and other carrier agents that may aid or increase the viral entry into the target site (such as for example a lipid delivery system).
  • the pharmaceutical compositions can be administered by any one or more of: inhalation, in the form of a suppository or pessary, topically in the form of a lotion, solution, cream, ointment or dusting powder, by use of a skin patch, orally in the form of tablets containing excipients such as starch or lactose, or in capsules or ovules either alone or in admixture with excipients, or in the form of elixirs, solutions or suspensions containing flavouring or colouring agents, or they can be injected parenterally, for example intracavemosally, intravenously, intramuscularly or subcutaneously.
  • compositions may be best used in the form of a sterile aqueous solution which may contain other substances, for example enough salts or monosaccharides to make the solution isotonic with blood.
  • compositions may be administered in the form of tablets or lozenges which can be formulated in a conventional manner.
  • the delivery of one or more therapeutic genes by a vector system according to the invention may be used alone or in combination with other treatments or components of the treatment.
  • Diseases which may be treated include, but are not limited to: cancer, neurological diseases, inherited diseases, heart disease, stroke, arthritis, viral infections and diseases of the immune system.
  • Suitable therapeutic genes include those coding for tumour suppressor proteins, enzymes, pro-drug activating enzymes, immunomodulatory molecules, antibodies, engineered immunoglobulin-like molecules, fusion proteins, hormones, membrane proteins, vasoactive proteins or peptides, cytokines, chemokines, anti-viral proteins, antisense RNA and ribozymes.
  • a gene encoding a pro-drug activating enzyme is delivered to a tumour using the vector system of the invention and the individual is subsequently treated with an appropriate pro-drug.
  • pro-drugs include etoposide phosphate (used with alkaline phosphatase Senter et al., 1988 Proc. Natl. Acad. Sci. 85: 4842-4846); 5-fluorocytosine (with Cytosine deaminase Mullen et al. 1994 Cancer Res.
  • Doxorubicin-N-p-hydroxyphenoxyacetamide with Penicillin-V-Amidase (Kerr et al. 1990 Cancer Immunol. Immunother. 31: 202-206); Para-N-bis(2-chloroethyl) aminobenzoyl glutamate (with Carboxypeptidase G2); Cephalosporin nitrogen mustard carbamates (with b-lactamase); SR4233 (with P450 Reducase); Ganciclovir (with HSV thymidine kinase, Borrelli et al. 1988 Proc. Natl. Acad. Sci.
  • FIG. 1 shows the structure of transcription units from plasmids pESP, pONY3 and pONY2.lnlsLacZ.
  • FIG. 2 shows a PCR analysis of integrated EIAV vector. PCR was performed with either genomic DNA from EIAV vector transduced cells (lanes 1 and 5) or mock transduced cells (lanes 2 and 6). pONY2.lnlsLacZ (lanes 3 and 7) and pONY3 (lanes 4 and 8) were used as controls.
  • FIG. 3 shows the structure of vector transcription units in deletion plasmids used to identify the packaging requirements for an EIAV vector.
  • FIG. 4 shows a secondary structure prediction for the RNA derived from the gag-transcription unit present in pONY2.13LacZ.
  • FIG. 5 is a representation of vectors derived from the EIAV genome.
  • FIG. 6 is a representation of gagpol constructs derived from EIAV.
  • FIG. 7 is a representation of an EIAV vector comprising an S2 deletion
  • FIG. 8 is a representation of EIAV gagpol constructs having deleted S2 and dUTPase genes.
  • FIG. 9 is a representation of an EIAV minimal vector.
  • FIG. 10 illustrates a gel showing an analysis of EIAV gagpol constructs according to the invention.
  • FIG. 11 shows examples of the pONY4 vectors.
  • FIG. 12 shows two SIN vectors.
  • FIG. 13 is a representation of a vector with a split polyA signal.
  • FIG. 14 is a representation of a vector with a split polyA signal.
  • FIG. 15 is a representation of a vector with a split polyA signal.
  • FIG. 16 shows construction of pONY4-GFP with a split polyA signal.
  • FIG. 17 shows construction of a MLV/EIAV vector.
  • FIG. 18 shows primers for construction of MLV/EIAV vectors.
  • FIG. 19 shows complete sequence of pONY-mouse.
  • FIGS. 20 and 21 give PCR priming.
  • FIG. 22 shows pEMVA4 (after PCR with primers EMVA 1-8).
  • FIG. 23 shows pEMVA4.
  • FIG. 24 shows pEMVA5.
  • FIGS. 25 and 26 show an example of hammer-head strategy for 5′ end formation.
  • FIG. 27 shows pEMVA6.
  • FIG. 28 shows pEMVA7 and pSyn pONY4.1.
  • FIG. 29 shows EMVA 10/11.
  • FIG. 30 shows pEMVA9.
  • FIG. 31 shows pEMVA10.
  • FIG. 32 shows pLWHORSE3.1.
  • FIG. 33 shows pMCRev.
  • FIG. 34 shows pYFVSVG.
  • FIG. 35 shows pYFAmpho.
  • FIG. 36 shows recombinant MVA constructs.
  • FIG. 37 shows the complete sequence of pSC65.
  • FIG. 38 shows the complete sequence of pLW22
  • FIG. 1 Plasmids used in this study. The genomic organization of EIAV is indicated including splice donor (d1, d2 and d3) and splice acceptor sites (a1, a2 and a3). The positions of gag, pol, env, tat, rev, S2 and the viral LTRs are also shown. Plasmid pESP is an EIAV vector genome containing the SV40 promoter and the puromycin resistance gene. Plasmid pONY3 is an EIAV gagpol expression plasmid. pONY2.lnlslacZ is an EIAV vector genome containing a HCMV IE enhancer/promoter and ⁇ -galactosidase gene (nlslacZ).
  • FIG. 2 shows PCR analysis of integrated EIAV vector. PCR was performed with either genomic DNA from EIAV vector transduced cells (lanes 1 and 5) or mock transduced cells (lanes 2 and 6). pONY2.lnlsLacZ (lanes 3 and 7) and pONY3 (lanes 4 and 8) were used as controls.
  • FIG. 10 shows analysis of gagpol expression constructs. 30%lg of total cellular protein was separated by SDS/Page electrophoresis, transferred to nitro-cellulose and probed with anti-EIAV antibodies. The secondary antibody was anti-Horse HRP (Sigma). Titres were averaged from three independent experiments and calulated as lacZ forming units per ml. There was no more than 10% variation between experiments. pONY2.lnlslacZ and the envelope expression plasmids were co-transfected with the EIAV gagpol.
  • VSV-G vesicular stomatitis virus glycoprotein
  • the resulting supernatant was used to transduce human kidney (293T) and canine osteosarcoma cells (D17) as follows. 48 hours post-transfection tissue culture fluid was collected and filtered through 0.45 cm filters. Ten-fold dilutions were made in culture medium containing polybrene at 8 ⁇ g/ml and then 500 ⁇ l aliquots placed on D17 cells seeded at 1.6 ⁇ 10 5 /well in 12 well plates on the previous day. Two hours later 1 ml of culture media was added. Two days later puromycin was added to a final concentration of 4 ug/ml and incubation was continued for a further 7 days.
  • a Murine leukaemia virus (MLV) based vector (pTIN500) containing the puromycin resistance gene under the control of the SV40 early promoter was used in conjunction with pHIT60 (MLV gagpol) and pRV67 (Cannon et al 1996 J. Virol. 70:8234-8240). No resistance colonies were detected on either cell type after 7 days of puromycin selection with the EIAV vector.
  • the MLV vector produced 5.0 ⁇ 10 4 c.f.u./ml on 293T cells and 1.0 ⁇ 10 4 c.f.u./ml on D17 cells.
  • a further vector system was therefore constructed comprising three transcription units to produce the following: 1) vector genome RNA; 2) env and 3) gag-pol.
  • the env and gag-pol transcription units are transcribed from a promoter-enhancer active in the chosen human packaging cell line. In this way, sufficient gag-pol and, most likely tat, are produced to ensure efficient production of transduction-competent vector particles.
  • the vector genome was constructed which has the reporter gene within the pol region of the genome as follows.
  • the plasmid designated pONYl was constructed by inserting the EIAV LTR, amplified by PCR from pSPEIAV19, into pBluescript II KS+(Stratagene).
  • the 5′ LTR of EIAV clone pSPEIAV19 was PCR amplified using pfu polymerase with the following primers:
  • the amplicon was blunt ended by 5′ overhang fill-in and inserted into pBluescript II KS+cut with Bss HII which had been blunt ended by 3′ overhand removal using T4 DNA polymerase.
  • This construct was called pONY1 and the orientation was 5′ to 3′ in relation to ⁇ -galactosidase of pBluescript II KS+. Sequencing of pONY1 revealed no mutations.
  • Vector genome pSPEIAV19DH was cut with Mlu I (216/8124) and inserted into pONY1 Mlu I cut (216) to make pONY2.
  • a Bss HII digest (619/792) of pBluescript II KS+ was carried out to obtain the multiple cloning site. This was blunt ended by 5′ overhang fill-in and ligated to pONY2 cut with Bgl II and Nco I (1901/4949) and blunt ended by 5′ overhang fill-in. The orientation was 3′ to 5′ in relation to the EIAV sequence. This was called pONY2.1.
  • pSPCMV was created by inserting pLNCX (Accession number: M28246) (Pst I/Hind III) into pSP72 (Promega).
  • the ⁇ -galactosidase gene was inserted from pTIN414 (Cannon PM et al J. Virol. 70, 8234-8240) into pSP72 (Xho I/Sph I) to make pSPlacZ.
  • the 5′ end to the ⁇ -galactosidase gene was replaced by the SV40 T antigen nuclear localization signal from pAD.RSVbgal (J. Clin. Invet. 90:626-630, 1992).
  • pAD.RSVbgal was cut with Xho I/Cla I and inserted into Xho I/Cla I pSPlacZ to make pSPnlslacZ.
  • the CMV nuclear localizing and non nuclear localizing ⁇ -galactosidase from pSPlacZ and pSPnlslacZ was cut out with Pst I and inserted into the Pst I site of pONY2.1 in the 5′ to 3′ orientation of EIAV. These were called pONY2. lnlslacZ and pONY2.1lacZ.
  • EIAV gagpol expression plasmid (pONY3) was then made by inserting Mlu I/Mlu I fragment from pONY 2 ⁇ H into the mammalian expression plasmid pCl-neo (Promega) such that the gag-pol gene is expressed from the hCMV-MIE promoter-enhancer.
  • gagpol pSPEIAV19DH was cut with Mlu I (216/8124) and inserted into pCI-Neo (Promega) Mlu I cut (216) to make pONY3. Plasmid pONY3 should not produce a functional genome because it lacks the appropriate LTR sequences.
  • Virus was produced by transient three plasmid cotransfection of 293T cells with pRV67, pONY3 and pONY2.10nlsLacZ as described for MLV-based vectors (Soneoka et al 1995 Nucl. Acids Res. 23:628-633) and then used to transduce 293T cells and D17 cells as follows. 48 hours post-transfection tissue culture fluid was collected and filtered through 0.45,um filters. Ten-fold dilutions were made in culture medium containing polybrene at 8 ⁇ g/ml and then 500 ul aliquots placed on D17 cells seeded at 1.6 ⁇ 10 5 /well in 12 well plates on the previous day.
  • the viral supernatants were used to transduce a variety of cell lines including human kidney (293T), murine embryo (NIH3T3) and canine osteosarcoma (D17).
  • the cell tropism of the virus was largely determined by the envelope.
  • EIAV could be pseudotyped with amphotropic envelope, but transduction efficiencies varied.
  • the amphotropic pseudotyped virus gave titres of about 102 on D17 cells, 103 on NIH3T3 cells and 104 on 293T cells. The reason for these differences was not pursued.
  • EIAV could also be pseudotyped with the MLV ecotropic envelope and these viruses transduced NIH3T3 cells at titres of 104 l.f.u./ml.
  • EIAV pseudotyped with VSV-G envelope
  • the titer varied between the different envelopes and cell types but overall efficiencies were relatively high for the non-murine cells, but still lower than with a murine vector system.
  • these data show that the EIAV vector system is not dependent on the EIAV envelope and can be effectively pseudotyped with three envelopes conferring broad host range. This makes this system as generally useful as current MLV-based systems.
  • EIAV vectors can also be pseudotyped in the same manner using the RD114 envelope, for instance using pRDF (Cosset et al 1995 J. Virol. 69: 7430-7436).
  • pONY2.10nlsLacZ was used as a positive control (FIG. 2A, lane 3).
  • No PCR product was detected when pONY3 was used as a template (FIG. 2A, lane 4).
  • RNA secondary structure prediction (“http://www.ibc.wustl.edu/ ⁇ zuker/rna/”) was used to identify possible stem-loop structures within the leader and the 5′ end of gag. Based on these predictions four deletions were made within the gag region of pONY2.10lacZ (FIG. 1). Deletions were made by PCR mutagenesis using standard techniques.
  • pONY2.1lacZ contains 1377 nt of gag (deleted from position 1901 nt)
  • pONY2.11lacZ contains 354 nt of gag (deleted from position 878 nt)
  • pONY2.12lacZ contains 184 nt of gag (deleted from position 708 nt)
  • pONY2.13lacZ contains 109 nt of gag (deleted from position 633 nt)
  • pONY2.14lacZ contains 2 nt of gag (deleted from position 526 nt)
  • gag coding sequence was needed for maximal packaging in addition to the un-translated region; pONY2.13lacz (Table 2). Similar titres were found on D17 cells.
  • the predicted secondary structure of the gag sequence derived RNA in pONY2.13lacZ is shown in FIG. 4.
  • pONY2.2lacZ contains deleted between position 409 to 421 nt
  • pONY2.22lacZ contains deleted between position 424 to 463 nt
  • pONY2.23lacZ contains deleted between position 470 to 524 nt
  • pONY2.24lacZ contains deleted between position 529 to 582 nt
  • pONY2.25lacZ contains deleted between position 584 to 645 nt
  • pONY2.26lacZ contains deleted between position 409 to 421 nt and between position 470 to 542 nt.
  • EIAV—based vector was made (pEGASUS-1) that contains only 759 nt of EIAV sequences (268 nt-675 nt and 7942 nt-8292 nt) as follows.
  • Sequences encompassing the EIAV polypurine tract (PPT) and the 3′LTR were obtained by PCR amplification from pONY2.10LacZ using primers PPTEIAV+(Y8198): GACTACGACTAGTGTATGTTTAGAAAAACAAGG, and 3′NEGSpeI (Y8199):CTAGGCTACTAGTACTGTAGGATCTCGAACAG.
  • the product was purified, digested with SpeI (ACTAGT) and ligated into pBS II KS+ which had been prepared by digestion with SpeI and treatment with alkaline phosphatase. Colonies obtained following transformation into E.
  • coli, XL-1Blue were screened for the presence of the 3′LTR in the orientation in which the U5 region of the 3′LTR was proximal to the NotI site of the pBS II KS+linker.
  • the sequence of the cloned insert was determined and showed that it contained only one change from the EIAV clone pSPEIAV19 (AC: U01866). This was a ‘C’ insertion between bases 3 and 4 of the R region. The same change was found in the template used in the PCR reaction.
  • the clone was termed pBS.3′LTR.
  • the reporter gene cassette CMV promoter/LacZ
  • the CMV/LacZ cassette was obtained as a PstI fragment from pONY2.10LacZ (see above).
  • the ligation reaction to join the above fragments was transformed into E. coli, XL-1Blue.
  • a number of clones in which the CMV/LacZ insert was orientated so that the LacZ gene was proximal to the 3′LTR were assessed for activity of the CMV/LacZ cassette by transfection into the cell line 293T using standard procedures.
  • a clone which gave blue cells at 48 hours post-transfection following development with X-gal was selected for further use and termed pBS CMVLacZ.3′LTR.
  • the 5′region of the EIAV vector was constructed in the expression vector pCIEneo which is a derivative of pCIneo (Promega) modified by the inclusion of approximately 400 base pairs derived from the 5′end of the full CMV promoter as defined previously.
  • This 400 base pair fragment was obtained by PCR amplifcation using primers VSAT1 (GGGCTATATGAGATCTTGAATAATAAAATGTGT) and VSAT2 (TATTAATAACTAGT) and pHFT60 as template.
  • VSAT1 GGGCTATATGAGATCTTGAATAATAAAATGTGTGT
  • VSAT2 TATTAATAACTAGT
  • pHFT60 pHFT60
  • a fragment of the EIAV genome running from the R region to nt 150 of the gag coding region was amplified with primers CMV5′EIAV2 (Z0591)(GCTACGCAGAGCTCGTTTAGTGAACCGGGCACTCAGATTCTG: and 3′PSI.NEG (GCTGAGCTCTAGAGTCCTTTTCTTTTACAAAGTTGG) using as template DNA.
  • the 5′region of the primer CMV5′EIAV2 contains the sequences immediately upstream of the CMV promoter transcriptional start site and can be cut with SacI.
  • 3′PSI.NEG binds 3′ of the EIAV packaging sequences as defined by deletion analysis (above) and contains an XbaI site.
  • the PCR product was trimmed with SacI and XbaI and ligated into pCIEneo which had been prepared for ligation by digestion with the same enzymes. This manipulation places the start of the EIAV R region at the transcriptional start point of the CMV promoter and transcripts produced thus start at the genuine start position used by EIAV and extend to the 3′-side of the packaging signal. Clones which appeared to be correct as assessed by restriction analysis were sequenced. A clone termed pCIEneo.5′EIAV was selected for further work.
  • the CMVLacZ and 3′LTR cassette in pBS.CMVLacZ.3′LTR was introduced into pCIEneo.5′EIAV.
  • pBS.CMVLacZ.3′LTR was digested with ApaI, the 3′ overhangs removed with T4 DNA polymerase, then digested with NotI.
  • the fragment containing the CMVLacZ.3′LTR was purified by standard molecular biology techniques.
  • the vector for ligation with this fragment was prepared from pCIEneo.5′EIAV by digestion with SalI, followed by filling-in of the 5′ overhangs using T4 DNA polymerase.
  • pEGASUS-1 The DNA was then digested with NotI and purified prior to use in ligation reactions. Following transformation into E. coli, XL-1Blue colonies were screened for the presence of the insert by restriction analysis to identify the required clone, designated pEGASUS-1.
  • EIAV vector pEGASUS-1 may be made by introduction of additional elements to improve titre.
  • a convenient site for the introduction of such elements is the SalI site which lies between the XbaI to the 3′ of the packaging signal and upstream of the CMV/LacZ cassette of pEGASUS-1.
  • the RRE from HV or EIAV can be inserted at this site.
  • the HIV-1 RRE was obtained from the HIV-1 molecular clone pWI3 (Kimpton and Emerman 1992 (J. Virol. 66: 2232-2239) by PCR amplification using primers RRE(+) GTCGCTGAGGTCGACAAGGCAAAGAGAAGAG and RRE( ⁇ ) GACCGGTACCGTCGACAAGGCACAGCAGTGG.
  • the fragment of DNA and pEGASUS-1 were digested with SalI and following ligation, transformed into E. coli, XL-1 Blue.
  • Colonies were screened for the presence of the HIV RRE and two clones, with the HIV RRE in either the positive or negative orientation, used for further work
  • vectors, pEGASUS-2.HIV RRE(+) or pEGASUS-2.HIV RRE( ⁇ ) can be tested in 293T cells by carrying out a four plasmid co-transfection in which the plasmid pCIneoHIVrev, expressing the rev protein from HIV-1 is co-transfected with vector, pONY3 and pRV67 plasmids
  • EIAV RRE as defined previously (Martarano et al 1994) was obtained by PCR amplification as follows. Using pONY2.10LacZ as template 2 amplifications were performed to obtain the two parts of the EIAV RRE.
  • the 5′-element was obtained using primers ERRE1 (TTCTGTCGACGAATCCCAGGGGGAATCTCAAC) and ERRE2 (GTCACCTTCCAGAGGGCCCTGGCTAAGCATAACAG) and the 3′element with ERRE3 (CTGTTATGCTTAGCCAGGGCCCTCTGGAAGGTGAC) and ERRE4 (AATTGCTGACCCCCAAAATAGCCATAAG).
  • the PCR amplification is set up with out primers ERRE1 and ERRE4 for the first 10 cycles and then these primers are added to the reaction and a further 10 cycles of amplification carried out.
  • the resulting PCR product and pEGASUS-1 were digested with SalI, ligated and transformed into E. coli XL-1Blue. Clones in which the EIAV RRE was in either the positive or negative orientations were selected for further work. The activity of these vectors was assessed in 4-way co-transfectioand pEGASUS-1 were digested with SalI, ligated and transformed into E.
  • EIAV REV encoding sequences were derived by PCR amplification.
  • the EIAV REV sequences were obtained using a two step ‘overlapping’ PCR amplification procedure as described above for the EIAV RRE.
  • Template for the two reactions was pONY3 and primers for the 5′fragment were EIAV REV REV 5′O(CCATGCACGTCTGCAGCCAGCATGGCAGAATCGAAG) and EAIV REV IN (CCTGAGGATCTATTTTCCACCAGTCATTTC) and for the 3′product EIAV REV IP (GTGGAAAATAGATCCTCAGGGCCCTCTGG) and EIAV.REV3′O (GCAGTGCCGGATCCTCATAAATGTTTCCTCCTTCG).
  • the second PCR amplification was carried out with primers EIAV REV5′O and EIAV REV3′O being added after 10 cycles.
  • the resulting product was ligated with the PCR fragment ‘TA’ cloning vector pCR2.1 (Invitrogen) the orientation of the EIAV REV insert was assessed by restriction enzyme analysis and the presence of the correct EIAV REV sequence confirmed.
  • the construct was called pTopoRevpos.
  • the EIAV REV insert was excised from pTopoRevpos by digestion with SpeI and NotI and ligated into pCIneo which had been digested with NheI and NotI.
  • PBMC peripheral blood mononuclear cells
  • Macrophages were obtained from this cell population by adherence to tissue culture plastic over 7 days in RPM! 1640 medium (Dutch modified; Sigma) containing 2% heat-inactivated human AB serum (Sigma) or 10% FCS (Sigma).
  • Non-adherent cells were removed by extensive washing of the plates with medium.
  • Virus for transduction experiments was obtained by three plasmid co-transfection into 293T cells.
  • the vector for the experiments was a pONY2.13 derivative in which the CMV/LacZ reporter cassette had been replaced with CMV/green fluorescent protein (GFP).
  • Vector pONY2.13GFP was made as follows. The sequence encoding the red-shifted GFP and eukaryotic translation signals was cut out of pEGFP-N1 (Clontech “http://www.clontech.com/”) with BglII and XbaI and ligated into the general cloning vector pSP72 (Promega) which had been prepared by digestion with the same enzymes. The GFP-encoding sequences were then excised using XhoI and ligated into pONY2.13 which had been cut with XhoI (thereby releasing the LacZ coding region). Following transformation into E. coli, XL-1Blue clones in which the orientation of the GFP insert with respect to the CMV promoter was such that expression would be expected were determined restriction analysis and expression of GFP confirmed by transfection of DNA into 293T cells.
  • Vector was recovered by three plasmid co-transfection into 293T cells and harvested at 42-48 hours post-transfection: tissue culture fluid was 0.45(m-filtered and virus was then pelleted by centrifugation at 50,000 g (20 Krpm), for 90 minutes at 4(C in a SW40Ti rotor. Virus was resupended in 50-100(1 of complete media for 2 hours and then used in transduction experiments. Transductions with pONY2.13GFP vector were carried out as follows. Macrophages, seeded at 5 ⁇ 10 5 per well of 48-well plates were washed once with medium and then 300 (l of medium was put back on the cells. Virus was added to the medium and gently pipetted up 2-3 times to ensure mixing.
  • Transduction efficiency was assessed at 3-5 days post-transduction.
  • the number of transduced macrophages was determined using a fluorescence microscope. Expression of GFP can be monitored for extended periods, e.g., up to several weeks.
  • transductions can be carried out with vectors carrying the LacZ marker. In such experiments the transduction frequency is assessed by detecting the presence of ⁇ -galactosidase using immunological procedures.
  • the brains were then dissected out, frozen into dry-ice cooled isopentane ( ⁇ 30° C.) and cut coronally at 10 ⁇ m with a cryostat. Every 5th section through the injection site and 2 mm rostral and caudal are collected onto Super-Frost slides, fixed and either X-gal or immunostained or stained with Cresyl Violet.
  • Epithelial cells can be differentiated to form epithelia-like monolayers which display (>1000 ⁇ cm 3 ) electrical resistance and a cuboidal morphology. There are various wasys to do this for example Fuller et al 1984.This creates polarized cells. This polarity is functional and mimics epithelial cells in vivo.
  • EIAV vectors can be used to transduce these cells either through the basolateral surface or the apical surface using vectors and preparations as described in Examples 1-3.
  • vector systems lacking tat, S2 and the dUTPase are constructed.
  • pONY2.13lacZ contains 109 nt of gag (deleted from nucleotide positions 633 to 4949) (pONY2.13lacZ is described above). This vector is used to make an EIAV vector genome from which S2 expression is eliminated by deletion from nucleotide positions 5345 to 5397. This removes the ATG start codon of S2 and the start codon of env. To make the deletion within S2, PCR is carried out with SY2/SY5 and SY3/SY4 using pONY2.13 DNA as template. The two PCR products are pooled and PCR is carried out with primers SY5 and SY3.
  • pONY2.13lacZDS2 is made by cutting out the 1.1 kb S2 region from pGEMS2 with Cel II and ligating it into pONY2.13lacZ.
  • pONY3DS2 is made by PCR amplification with SY1/SY2 and SY3/SY4 using pONY3 DNA as template.
  • the two PCR products are pooled and PCR is carried out with primers SY1 and SY3.
  • the 0.7 kb product is ligated into pGEMT-easy (Promega) to make pGEMS22 and sequenced to confirm the deletion.
  • the 0.7 kb S2 coding region is excised of pGEMS22 with Not I and inserted into pBluescript KS+(Stratagene) to make pBPCRS2.
  • the Eco RV and Nco I fragment from pONY3 (2.2 kb) is inserted into pBPCRS2 cut with Eco RV and Nco I to make pBpONYS2. This is then cut with Eco RV and Cel II (2.9 kb fragment) and inserted into pONY3 cut with Eco RV and Cel II to thereby making pONY3DS2.
  • pONY3DdUTPase is made by site directed mutagenesis of nucleotide 4176 from a T to an A residue (Payne et al., Virology, 210:302-313). This mutates the aspartic acid to a glutamic acid. This is done by PCR amplification using PCR primers dUTPaseF and dUTPaseR. The template DNA is pONY 3 . The PCR product is inserted into pGEMT-easy and sequenced to confirm the mutation. This is called pGDdUTPase.
  • pONY3 is cut with Not I and Eco RV (4.6 kb) and inserted into pBluescript KS+(Stratagene) to make pBEV.
  • the pGDdUTase is cut with Pac I and Pst I and the 0.4 kb band inserted into pBEV cut with Pac I and Pst I. This is called pONY3pBDUTPase.
  • This is then inserted into pONY3 via N ⁇ overscore (o) ⁇ t I and Eco RV (4.6 kb) to make pONY3DdUTPase.
  • pBpONYS2 is used.
  • pGDdUTPase is cut with Pac I and Pst I and the fragment inserted into pBpONYS2 cut with Pac I and Pst I to make construct pS2DdUTPase.
  • This is then cut with Eco RV and Cel II and inserted into pONY3 cut with Eco RV and Cel II to make pONY3DS2DdUTPase.
  • pONY2.13lacZDS2, pONY3DdUTPase and pONY3DS2DdUTPase vectors are used in a number of combinations in three plasmid co-transfections to generate virus as described for MLV-based vectors (Soneoka et al 1995 Nucl. Acids Res. 23:628-633) and the virus generated is titred on 293T and D17 cells, in either dividing or non-dividing states.
  • Cells are arrested in G 1 /S phase by treatment with aphidicolin (9) and then transduced with EIAV-based and MLV-based vectors pseudotyped with VSV-G (Table 4).
  • the transduction efficiency of the MLV vector is lower by four orders of magnitude in aphidicolin treated cells as compared to untreated cells.
  • the incomplete block to cell transduction by MLV is probably due to a small population of dividing cells.
  • no significant difference is observed in the case of the EIAV-based vectors.
  • the EIAV-based system does not require S2 or dUTPase either for production or transduction.
  • Payne et al. (Payne et al., Virology, 210:302-313) and others have shown that EIAV dUTPase is required for the infection of horse macrophages. This may represent a restriction in infection of macrophages by EIAV.
  • pONY3 In pONY3 there is an extended 5′ untranslated region before the start of the gagpol coding sequence. It is likely that this unusually long sequence would compromise expression of the gagpol cassette.
  • pONY3 is modified to remove the remaining 5′ LTR. This is done by cutting pONY3 with Nar I and Eco RV. The 2.4 kb fragment is inserted into pBluescript KS+(Stratagene) at Cla I and Eco RV sites to make construct pBSpONY3.0. pBSpONY3.0 is cut with Xho I and Eco RV. The 2.4 kb fragment is inserted into pONY3 at Xho I and Eco RV to make pONY3.1.
  • pONY3.1 like pONY3 encodes gag, gagpol, Tat, S2 and Rev. Since the S2 mutation experiments showed that S2 is not required either in the production system or in the EIAV vector genome it is possible to design a gagpol expression constructs without S2. Two such constructs, pHORSE and pHORSE3.1, are produced.
  • pHORSE is made by PCR amplification with EGAGP5′OUTERIEGAGPINNER3 and EGAGP3′OUTER/EGAGPINNER5 using pONY3 as template DNA.
  • the two PCR products are purified pooled and re-amplified using primers EGAGP5′OUTER/EGAGP3′OUTER.
  • This product is inserted into the Xho I and Sal I sites of pSP72 to make pSP72EIAVgagpolO'lap.
  • pONY 3 is cut with Pvu II and Nco I and the 4.3 kb fragment is inserted into pSP72EIAVgagpolO'lap cut with Pvu II and Nco I to make pSPEGP.
  • pHORSE3.1 is made by replacing the 1.5 kb Xho IXba I of pHORSE with the 1.6 kb Xho I/Xba I of pONY3.1. Titres obtained with pHORSE3.1 are similar to that of pONY3.1.
  • Codon optimisation of the EIAV gagpol should eliminate the dependence of gagpol protein expression on the RRE/Rev system.
  • the need of pEGASUS-1 for Rev/RRE can also be eliminated by using a heterologous RNA export system such as the constitutive transport element (CTE) from Mason-Pfizer Monkey virus (MPMV) (Bray et al., PNAS, 1994, 91:1256-1260, Kim et al., 1998) TABLE 4 Titre on Ratio D17 cells (Non- (l.f.u./ml) Non- dividing/ Vector gagpol Dividing dividing dividing) S2+ S2+, dUTPase+ 2.2 ⁇ 10 5 1.1 ⁇ 10 5 0.5 S2 ⁇ S2+, dUTPase+ 1.5 ⁇ 10 5 1.3 ⁇ 10 5 0.9 S2 ⁇ S2 ⁇ , dUTPase+ 1.0 ⁇ 10 5 1.2 ⁇ 10 5 1.2 S2 ⁇ S2 ⁇ , d
  • Titres with Rev are higher for pEGASUS-1 even though it has no RRE. Possibly the effect of REV is via enhanced expression of gagpol.
  • TABLE 6 Titre Vector Genome Gagpol (l.f.u./ml) pONY2.11lacZ pONY3.1 1.7 ⁇ 10 5 pONY2.11lacZ pHORSE3.1 9.0 ⁇ 10 4 pEGASUS/RRE pONY3.1 8.0 ⁇ 10 4 pEGASUS/RRE pHORSE3.1 2.0 ⁇ 10 4
  • pONY2.1lacZ contains a deletion in gag such that only 373 bp of the gag ORF remains.
  • pONY4 was made by replacing the 5′ LTR with the CMV LTR from pEGASUS-1.
  • pEGASUS-1 was cut with Bgl II/Xho I releasing a 3.2 kb fragment (containing the CMV LTR) which was inserted into pSP72 cut with Bgl I/Xho L This construct was named pSPPEG213.
  • pONY4.1 contains a deletion (20.1 kb) downstream of the lacZ gene (between the Sfu I and Sal I sites) such that tat, S2, env, rev and RRE, are either missing or severely truncated (FIG. 11 c ).
  • pONY4.1 was made by cutting it with Sfi I/Sal I, blunt-ended by Klenow polymerase and religated.
  • pONY4G was made by replacing the lacZ gene of pONY4 (Sac II/Kpn I and then blunting with Klenow polymerase) with that of GFP from pEGFP-N1 (Clontech) (Bam HI/Xba I and then blunting with Klenow polymerase) as a blunt fragment.
  • Vector stocks were generated by calcium-phosphate transfection of human kidney 293T cells plated on 10 cm dishes with 16 ⁇ g of vector plasmid, 16 ⁇ g of gag-pol plasmid and 8 ug of envelope plasmid. 36-48 h after transfection, supernatants were filtered (0.45,um) aliquoted and stored at ⁇ 70° C. Concentrated vector preparations were made by ultracentrifugation of at 20 000 rpm (SW40Ti rotor) for 90 min, at 4° C. The virus was resuspended in PBS for 3-4 h aliquoted and stored at ⁇ 70° C. Transduction was carried out in the presence of polybrene (8 ⁇ g/ml).
  • the expression of the transgene from EIAV vectors in particular cell types may be influenced by elements in the LTR's.
  • SIN Self Inactivating
  • RNA structure is a determinant of poly(A) site recognition by cleavage and polyadenylation specificity factor.
  • Graveley B R, Fleming E S, Gilmartin G M) J Virol 1996 Mar; 70(3):1612-7.
  • SIN vectors provide a way for eliminating the production of full length transcripts in transduced cells.
  • Two SIN vectors were made: one containing the putatively important sequences (for polyadenylation), located between the Mlu I and Mun I sites and one in which these sequences were deleted.
  • the 5′ border of the deletions was 112 bases from the 5′ end of the U3 region of the 3′LTR.
  • the structure of two SIN vectors is shown in FIG. 12.
  • DNA sequences between nucleotides 7300 and 8079 were obtained using polymerase chain reaction amplification using pONY4G as template.
  • the positive sense primer was ERRE3 and the negative primers for amplification were SIN-MLU (C7143: GTCGAGCACGCGTTTGCCTAGCAACATGAGCTAG (MluI site in bold) or SIN-MUN (C7142: GTCGAGCCAATTGTTGCCTAGC AACATGAGCTAG (MunI site in bold) where the underlined sequences are complimentary to nucleotides 8058 to 8079 (of pSPEIAV19).
  • PCR products were digested with NspV and either MuI or MunI respectively. These were then ligated into pONY4G prepared for ligation by digestion with NspV(SfuI) and either MluI (partial digestion) or MunI respectively.
  • EIAV vectors such as pONY4Z or pONY4G
  • the internal CMV-reporter cassette is orientated so that transcription from the 5′LTR and the internal promoter are co-directional and the polyadenylation signal in the 3′LTR is used for transcripts from both promoters.
  • An alternative configuration is achieved by reversing the internal promoter-reporter cassette, however a polyadenylation signal must be placed downstream of the cassette.
  • This ‘reverse orientation’ vector was made as follows. pONY4Z was digested with PstI and the overhanging termini trimmed back with T4 DNA polymerase. This was then used as the ‘vector’ fragment in a ligation with the MluI to AseI fragment from pEGFP-C1 which contains sequences including the CMV-GFP-SV40 early mRNA polyA signal cassette. Prior to ligation this fragment was flush-ended with T4 DNA polymerase. The vector encoding plasmid was called pONY4Greverse.
  • Vector particles were recovered from pONY4Greverse by co-transfection with pONY3.1 and pRV67, which express EIAV gag/pol and VSV-G protein respectively.
  • the titre on D17 canine cells from pONY4Greverse was 13-fold lower than from pONY4G vector recovered in parallel.
  • This ‘vector’ fragment was then ligated to a DNA fragment representing the BGHpA which was prepared from pcDNA3.1+ (Invitrogen) by digestion with SphI, and then the ends blunted with T4 DNA polymerase, then digested with PstI.
  • the polyadenylation signal used is that from EIAV. This is found in the 3′ LTR at the border of R and U5. This signal may not be optimal because it is not of a consensus sequence (see Whitelaw and Proudfoot 1986 EMBO 5; 2915-2922 and Levitt et al 1989 Gen. and Dev. 3; 1019-1025 for description of consensus polyadenylation signal).
  • One method of improving the viral polyadenylation is to replace the 3′ LTR poly A signal with that of a consensus/strong polyadenylation signal. By such a method the signal would now be optimal in the producer cell. However upon transduction this signal is lost because during replication, the 5′ LTR is the source of the poly A signal (see Retroviruses 1998 CSH press (Ed. J. Coffin) for review of retroviral life cycle).
  • One novel way of overcoming the problem is to include the poly A signal in a manner as will now be outlined: The method is to use a ‘split poly-A signal’ where by an intron splits the aataaa motif from that of the essential g/u box.
  • Such a signal has previously been used by Liu et al (1993 N.A.R 21;5256-5263) to demonstrate both that large gaps between the aataaa and the g/u box will disable the poly A signal and that the polyadenylation process preceeds splicing.
  • a split-polyA signal within the retroviral vector such a signal will not be functional until transduction of target cells. This is because polyadenylation preceeds splicing and as such the upstream split-polyA signal will not be used during vector expression within the producer cell.
  • Outlined in FIG. 13 is a schematic representation of how such a retroviral vector, containing a split polyA signal, would function—both in producer and in transduced cells.
  • this Figure demonstrates that although there exists an upstream consensus polyadenylation signal, the initial vector transcripts are still polyadenylated at the usual 3′ LTR using either a viral or other poly A signal as so desired. This is because although the upstream poly A signal is functional in the final vector genome, this signal is not read by the polyadenylation machinery because it is created only during intron removal and thus not present in the primary RNA transcript. Second, this figure demonstrates that upon transduction the-resulting vector transcripts are now polyadenylated at the first signal; this being now a normal strong polyadenylation signal with no introns to distance the essential aataaa and g/u box.
  • the viral poly A signal in the 5′ LTR (the one copied to the 3′ position during reverse transcription) can be removed/deleted if desired. This is of use for preventing the process of promoter-proximal polydenylation from the 5′ LTR in the producer cell (see Scott and Imperiale 1997 (Mol. Cell. Biol. 17;2127-35) and thus encourage full length transcript production of the virus.
  • the “split poly A signal” cassette is constructed as described in FIG. 15; with the intronic sequence being derived from pCI (Promega). Once made this cassette is cloned into the pONY 4 GFP vector using the PstI compatible unique sse8387 site of pONY4-GFP (see FIG. 16). Upon transduction the resulting vector will now polyadenylate prior to the 3′LTR and consequently no viral RNA 3′ to lacZ will be transcribed (see FIG. 16).
  • the final full length product is cut BglII and Xbal and used to replace the BglII-Xbal fragment of pONY4 lacZ (containing the CMV/R/U5) to make pONY4-5′MLV.
  • the resulting vector now has the CMV/R/U5 sequence from MLV linked to the EIAV U5 sequence (sequence required for genome recognition by intergrase prior to intergration).
  • the next step involves PCR amplification with primers Me5 and Me6 from pONY4 LacZ template and PCR amplification with Me7 and Me8 from pLXSN template (Miller and Rosman 1989 Biotechniques 7:980-990).
  • PCR products are then overlapped by primerless PCR (homology between primers shown as hatched box) and the resulting fragment cut with NspV and MunI and inserted into the NspV/MunI sites of pONY4-5′MLV; thus replacing the 3′EIAV LTR with a 3′MLV LTR fused to the 3′UTR/ppt/U3 intergrase binding site of pONY 4 lacZ.
  • the resulting plasmid named pONY-MOUSE (see FIG. 19 for complete DNA sequence), titres at 10 4-5 per ml when combined with pONY3.1 and pRV67 in the HIT system.
  • an EIAV genome is expressed from a vaccinia early promoter P7.5E (Davison 1989a).
  • the promoter has been engineered to produce an EIAV genome with the correct 5′ RNA end.
  • the vaccinia early termination sequence has been inserted downstream of the EIAV genome. This is inserted into the transfer plasmid pSC65, which can homologously recombine into the TK region of the MVA genome. Recombinant viruses can be selected by their lack of sensitivity to BudR (Earl et al. 1998).
  • FIG. 11 is a schematic representation of the EIAV genome vectors pONY4.0 and pONY4.1 which have been described in Example 10 and the vaccinia transfer vector pSC65 (Chakrabarti et al 1997).
  • the P7.5E sequence is AAAAGTAGAAAATATATTCTAATTTATT.
  • FIGS. 20 and 21 give the sequence of the PCR primers.
  • PCR with primers EMVA1/2 produces the 5′ LTR with the U3 region replaced by the P7.5E promoter. This is inserted into the plasmid pSP72 (Promega) using the Hind III/Pst I sites to make pEMVA1.
  • EIAV U3 contains a sequence matching the criteria for vaccinia early termination (TTTTTAT). Using primers EMVA3/4 and EMVA5/6 and overlapping PCR this region is mutated to TTTCCAT in order to prevent early termination.
  • This PCR product is inserted into the pEMVA1 using the Bgl II/Pst I sites to generate pEMVA2.
  • TTTTTTTTTTT is inserted downstream of the 3′ LTR R region using primers EMVA7/8 .
  • This PCR product is inserted into pEMVA2 using the Mun I/Bgl II sites making pEMVA3.
  • pONY4 is inserted via the Nar I/Nsp V sites making pEMVA4 (FIG. 22).
  • This is then cut with Pac I/Bgl II and inserted into pSC65 cut with Pac I/Bam HI to make pEPONY4 (Bgl II and Bam HI are compatible) (FIG. 23). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65.
  • pEMVA4 is cut with Sal I/Nsp V blunt ended and religated to make pEMVA5 (FIG. 24). This removes much of the sequence between the end of the lac Z gene and the end envelope region, hence this vector is Tat, Rev, S2 and Env minus. This is described in Example 10. This is then cut with Pac I/Bgl II and inserted into pSC65 cut with Pac I/Bam HI to make pEPONY4.1 (Bgl II and Bam HI are compatible) (FIG. 24). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65.
  • Both pEPONY4.0 and pEPONY4.1 are suitable for inserting the genome expression cassettes into the TK region of the MVA genome (Carroll MW and Moss B Virology Nov. 24, 1997;238(2):198-211) using a BHK TK-ve cell line (ECACC 85011423) and standard procedures for the construction of recombinant poxviruses (Earl et al 1998a & 1998b)
  • the synthetic early/late promoter of vaccinia has a requirement for sequences downstream of the RNA initiation site (Davison 1989b).
  • this promoter For this promoter to be used to generate a retroviral genome either the R regions have to be modified or a ribozyme is used to make the correct 5′ end. Modifying the R regions is problematic as the initiation site has not been conclusively identified and varies with the sequence (Davison 1989b).
  • a transfer plasmid that expresses the EIAV genome from the synthetic early/late promoter (Psyn). Downstream of this promoter is inserted a ribozyme that ensures the creation of the correct 5′ end of the RNA. This construct also contains the early termination sequence.
  • PCR with primers EMVA9/1 produces the 5′ LTR with the U3 region replaced by the Psyn promoter and a hammerhead ribozyme (FIGS. 25 and 26).
  • This is inserted into the plasmid pEMVA4 (Example 15) using the Pac I/Nar I sites to make pEMVA6 (FIG. 27).
  • This is then cut with Pac I/Bgl II and inserted into pSC65 cut with Pac I/Bam HI to make pSynPONY4 (Bgl II and Bam HI are compatible) (FIG. 27). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65.
  • pEMVA6 is cut with Sal I/Nsp V blunt ended and religated to make pEMVA7 (FIG. 28). This is then cut with Pac I/Bgl 11 and inserted into pSC65 (a vaccinia transfer vector) cut with Pac I/Bam HI to make pSynPONY4.1 (Bgl II and Bam HI are compatible) (FIG. 28). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65.
  • Both pSynPONY4.0 and pSynPONY4.1 are suitable for inserting the genome expression cassettes into the TK region of the MVA genome (Carroll MW and Moss B Virology Nov. 24, 1997;238(2):198-21 1) using standard procedures for the construction of recombinant poxviruses (Earl et al 1998a & 1998b)
  • the T7 promoter can be used to generate a retroviral genome which can make the correct 5′ end.
  • T7 T7 promoter
  • T7 termination sequence T7
  • the T7 promoter requires the T7 polymerase.
  • MVA viruses are available which express T7 polymerase from Vaccinia promoters (Wyatt et al 1995).
  • the T7 promoter has the sequence ( ⁇ )TAATACGACTCACTATAGG(+2) with transcription beginning after A with preferably a run of Gs.
  • the T7 termination sequence is CTAGCATAACCCCTTGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTG.
  • the T7 promoter and terminator sequences are as those described in the plasmid pCITE-4a(+) (Novagen).
  • PCR with primers EMVA10/11 (FIG. 29) produces the 5′ LTR with the U3 region replaced by the T7 promoter. This is inserted into the plasmid pEMVA4 using the Pac liNar I sites to make pEMVA8 (FIG. 30). PCR with primers EMVA 1/7 produces part of the 3′ LTR with a T7 termination sequence. This is inserted into pEMVA8 using the Mun I/Bgl II sites to make pEMVA9.
  • pEMVA9 is cut with Sal I/Nsp V blunt ended and religated to make pEMVA10 (FIG. 31). This is then cut with Pac I/Bgl II and inserted into pSC65 (a vaccinia transfer vector) cut with Pac I/Bam HI to make pT7PONY4.1 (Bgl II and Bam HI are compatible) (FIG. 31). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65.
  • Both pT7PONY4.0 and pT7PONY4.1 are suitable for inserting the genome expression cassettes into into the TK region of the MVA genome (Carroll MW and Moss B Virology 1997) using standard procedures for the construction of recombinant poxviruses (Earl et al 1998a & 1998b).
  • EIAV gag/pol requires Rev/RRE for expression as Rev enables the unspliced transcript to be exported out of the nucleus.
  • Rev has other functions such as RNA stability or acts as a translation enhancer it can be expressed in a similar way to EIAV gag/pol (Martarano 1994).
  • Rev has other functions such as RNA stability or acts as a translation enhancer it can be expressed in a similar way to EIAV gag/pol (Martarano 1994).
  • the EIAV gag/pol sequence can be codon optimised to overcome the Rev/RRE requirement for export and enhance RNA stability. Below is described the creation of a vector that expresses EIAV gag/pol from a synthetic early/late promoter (Psyn).
  • EIAV gag/pol including the leader the gag/pol open reading frame and the RRE can be obtained from cutting pHORSE3.1 (Example 9) with Aho LINot I to give a 5.5 kb band (FIG. 32). This is then inserted into the vaccinia transfer vector pLW-22 cut with Sal I/Not I (Sal I and Xho I are compatible) to make pLWHORSE3.1 (FIG. 32).
  • Rev In the event that Rev is required for EIAV viral vector production from a poxvirus it can be expressed from a synthetic early/late promoter. This construct is inserted into the transfer plasmid pMC03, which can homologously recombine into the Del III region of the MVA genome. Recombinant viruses can be selected by their expression of GUS (Carroll et al. 1995).
  • Plasmid pCIRev is described in Example 9. It is an EIAV Rev expression plasmid. This is cut with Afl II/Not I (0.6 kb), blunt ended by T4 DNA polymerase and inserted into pMC03 (Carroll et al. 1995) cut with Pme I to make pMCRev (FIG. 33).
  • EIAV can be pseudotyped with a number of envelopes such as VSV-G and amphotropic MLV envelope.
  • VSV-G a number of envelopes
  • amphotropic MLV envelope a number of envelopes such as VSV-G and amphotropic MLV envelope.
  • the transfer vector is pYF6 which can homologously recombine into the HA region of MVA.
  • Recombinant viruses can be selected by direct live immunostaining for expression of the env.
  • VSV-G expression plasmid pRV67 (Kim et al. 1998) is cut with Sma I/Eco RV(1.7 kb) and the resulting fragment inserted into pYF6 cut with Sma I to make pYFVSVG (FIG. 34).
  • pHIT456 (Soneoka 1995) is cut with Xba I and the 2.2 kb band blunt ended by T4 DNA polymerase and inserted into pYF6 cut with Sma Imaking pYFAmpho (FIG. 35).
  • Both pYFAmpho and pYFVSVG are suitable for inserting the genome expression cassettes into into the HA region of the MVA genome using standard procedures for the construction of recombinant poxviruses (Earl et al 1998a & b, Flexner et al 1987)
  • the recombinant vaccinia viruses containing multiple inserts encoding the components of the EIAV vectors are constructed by sequential recombination with the relevant transfer plasmids.
  • the construction of v.MEeG-Or is used as an example:
  • a plasmid carrying gag-pol (pLWHORSE3.1) is transfected into BHK-21 or CEF cells, that have been previously infected with MVA (as described in Carroll and Moss 1997, Earl et al 1998a & b).
  • Recombinant virus is then used to recombine with transfer plasmids containing the other recombinant genes: pMCRev in which selection is based on GUS expression (Carroll & Moss 1995), the genome (pEPONY4.0) in which selection is based on a TK negative phenotype using BudR (Carroll & Moss 1997, Earl et al 1998a & b) and VSVG (PYFVSVG) in which recombinants are identified by direct immunostaining of VSV G (Earl et al 1998a & b).
  • Recombinant viruses may be amplified in BHK-21 or CEF cells as described below:
  • the highly attenuated strain MVA is derived from the replication competent strain Ankara and has endured over 570 passages in primary chick embryo fibroblast cells. MVA replication was initially thought to be restricted to CEF cells as only minimal replication in mammalian cells was reported. However, further analysis has shown that Baby Hamster Kidney cells (BHK-21) are able to support high titre production of MVA. MVA may thus be grown on BHK-21 or primary CEF cells (Carroll & Moss (1997) Virology 238:198-211).
  • CEF cells 10 day old chick embryos are gutted and limbs and head are removed before being minced and trypsinised in a solution of 0.25% trypsin and incubation at 37° C. The cell suspension is filtered through a course filter mesh and cells are washed and concentrated by centrifugation at 2000 rpm in a Sorvall RC-3B at 1500 rpm for 5 mins. Cells are suspended in MEM containing 10% FCS, aliquotted into 175 cm flasks and incubated at 37° C. in a CO 2 incubator. When monolayers are 95% confluent they are trypsinised and used to seed additional flasks or six well plates. Alternatively, primary cultures are transferred to a 31° C. incubator for later use (Sutter and Moss (1992) Proc Natl Acad Sci U S A 89:10847-10851).
  • Crude virus stocks are prepared for initial recombinant virus analysis or as viral stocks used for subsequent high titre virus preparations.
  • Vaccinia virus preparations can be semi-purified by centrifuging out cell membranes and nuclei or by additional steps involving sucrose centrifugation to prevent contamination by pre-expressed recombinant protein products and cellular organelles. Methods used are a modification of those described by Earl et al., 1998a & b; Earl and Moss, ibid, pp. 16.17.1-16.17.16; Earl and Moss, ibid, pp. 16.18.1-16.18.10; and Bronte et al., (1997) Proc Natl Acad Sci U S A 94(7):3183-3188.
  • MVA is grown in either CEF or BHK-21 (obtained from the ATCC) and WR is grown in HeLa or BS-C-1 (ATCC) in 175 cm 2 tissue culture flasks. Briefly, confluent monolayers are infected with an moi of approx. 1 pfu with MVA or WR. Virus is suspended in 10 ml MEM containing 2% FCS and added to 175 cm 2 flasks containing confluent cell monolayers. After inoculation for 1 hour at 37° C. an additional 20 ml MEM containing 2% FCS is added. After 48-72 hours infected cells are scraped into the medium and pelleted at 1500 g for 5 mins.
  • Virus stocks are stored below ⁇ 20° C.
  • Infected cells are harvested as described previously (Earl et al a & b; Earl and Moss; 1991). After centrifugation cells are resuspended in PBS (2 ml/175 cm 2 flask) and homogenised by 30-40 strokes in a tight fitting glass dounce homogeniser, on ice. Cell breakage is checked by microscopy. Nuclei, cellular organelles and membranes are removed by a centrifugation at 300 g for 5 mins (4° C.), keep supernatant. The cell pellet is resuspended in 1 ml/175 cm 2 flask and centrifugation repeated. The supernatants are pooled, aliquoted and stored.
  • Infected cells are harvested as previously described (Earl et al.a & b; Earl and Moss; 1991) and resuspended in 10 mM Tris.Cl, pH 9.0 (2 ml/flask), keeping samples on ice from this point of the procedure. Homogenise as described previously using 10 mM Tris. The lysate is sonicated (on ice) using an XL 2015 sonicating cup (Misonics, USA) at maximum output (500 W) for 1 min. The sample is placed on ice for 1 min and the sonication repeated up to 3 times. A maximum of 5 ml is sonicated at a time, and ice is replenished during sonication.
  • the lysate is gently layered onto a cushion of 17 ml of 36% sucrose (in 10 mM Tris.Cl, pH 9.0) in a SW-27 centrifuge tube. Lyates are centrifuged for 80 mins in an SW-27 rotor at 13 500 rpm (32,900 ⁇ g), 4° C. The supernatant is discarded and the viral pellet resuspended in sterile PBS and sonicated in a cup sonicator for 1 min (on ice). Concentrated virus is aliquoted and stored at below ⁇ 20° C.
  • MVA-EIAV large scale preparations of recombinant MVA-EIAV are made. These preparations are used to infect mammalian cells that are non-permissive for MVA, such that the resulting supernatant will only contain EIAV and not infectious MVA (Meyer et al 1991, Carroll and Moss 1997).
  • a suitable cell line is MRC5 (ATCC). Cells are infected at an MOI of 3. Infections are allowed to run for approximately 48 hours before supernatants are harvested and EIAV vector particles either used directly or concentrated/purified by ultracentrifugation or cross-flow methods.
  • EIAV vectors carrying gene of interest prepared in these ways are used to transduce target cells in vivo or in vitro.
  • Equine infectious anemia virus trans-regulatory protein Rev controls viral mRNA stability, accumulation, and alternative splicing. J Virol 68, 3102-11.
  • Cannon 1996 J Virol 1996 70:8234-40 Murine leukemia virus-based Tat-inducible long terminal repeat replacement vectors: a new system for anti-human immunodeficiency virus gene therapy.
  • Moss B Poxviridae The viruses and their replication Chapter 83. p2637-2672.
  • Panicali D Construction of poxviruses as cloning vectors: insertion of the thymidine kinase gene from herpes simplex virus into the DNA of infectious vaccinia virus. Proc Natl Acad Sci U S A 1982 79:4927-31

Abstract

A retroviral vector derived from a non-primate lentivirus genome comprising a deleted gag gene wherein the deletion in gag removes one or more nucleotides downstream of nucleotide 350 of the gag coding sequence.

Description

  • This invention relates to a retroviral vector. In particular, but not exclusively, it relates to retroviral vectors capable of transferring genetic material to non-dividing or slowly-dividing cells derived from non-primate lentiviruses. [0001]
  • There has been considerable interest, for some time, in the development of retroviral vector systems based on lentiviruses, a small subgroup of the retroviruses. This interest arises firstly from the notion of using HIV-based vectors to target anti-HIV therapeutic genes to HIV susceptible cells and secondly from the prediction that, because lentiviruses are able to infect non-dividing cells (Lewis & Emerman 1993 J. Virol. 68, 510), vector systems based on these viruses would be able to transduce non-dividing cells (e.g. Vile & Russel 1995 Brit. Med. Bull. 51, 12). Vector systems based on HIV have been produced (Buchschacher & Panganiban 1992 J. Virol. 66, 2731) and they have been used to transduce CD4+cells and, as anticipated, non-diving cells (Naldini et al, 1996 Science 272, 263). In addition lentiviral vectors enable very stable long-term expression of the gene of interest. This has been shown to be at least three months for transduced rat neuronal cells. The MLV based vectors were only able to express the gene of interest for six weeks. [0002]
  • HIV-based vectors produced to date result in an integrated provirus in the transduced cell that has HIV LTRs at its ends. This limits the use of these vectors as the LTRs have to be used as expression signals for any inserted gene unless an internal promoter is used. The use of internal promoters has significant disadvantages. The unpredictable outcome of placing additional promoters within the retroviral LTR transcription unit is well documented (Bowtell et al, 1988 J. Virol. 62, 2464; Correll et al, 1994 Blood 84, 1812; Emerman and Temin 1984 Cell 39, 459; Ghattas et al, 1991 Mol. Cell. Biol. 11, 5848; Hantzopoulos et al, 1989 PNAS 86, 3519; Hatzoglou et al, 1991 J. Biol. Chem 266, 8416; Hatzoglou et al, 1988 J. Biol. Chem 263, 17798; Li et al, 1992 Hum. Gen. Ther. 3, 381; McLaichlin et al, 1993 Virol. 195, 1; Overell et al, 1988 Mol. Cell Biol. 8, 1803; Scharfman et al, 1991 PNAS 88, 4626; Vile et al, 1994 Gene Ther 1, 307; Xu et al, 1989 Virol. 171, 331; Yee et al, 1987 PNAS 84, 5197). The factors involved appear to include the relative position and orientation of the two promoters, the nature of the promoters and the expressed genes and any selection procedures that may be adopted. The presence of internal promoters can affect both the transduction titers attainable from a packaging cell line and the stability of the integrated vector. [0003]
  • HIV and other lentiviral LTRs have virus-specific requirements for gene expression. For example, the HIV LTR is not active in the absence of the viral Tat protein (Cullen 1995 [0004] AIDS 9, S19). It is desirable, therefore, to modify the LTRs in such a way as to change the requirements for gene expression. In particular tissue specific gene expression signals may be required for some gene therapy applications.
  • HIV vectors have a number of significant disadvantages which may limit their therapeutic application to certain diseases. HIV-1 has the disadvantage of being a human pathogen carrying potentially oncogenic proteins and sequences. There is the risk that introduction of vector particles produced in packaging cells which express HIV gag-pol will introduce these proteins into the patient leading to seroconversion. For these reasons, there is a need to develop lentiviral-based vectors which do not introduce HIV proteins into patients. [0005]
  • We have now found it possible to provide an improved lentiviral vector which overcomes the limitations of HIV-based vectors. It is important in the development of any retroviral vector system to remove sequences from the retroviral genome which may inhibit the capacity of the vector to transfer heterologous genes or which may transfer disadavantageous protein coding sequences to the target cell. Retroviruses are limited in the length of RNA sequences which can be packaged efficiently and so the existence of long regions of the retroviral genome severely limits the coding capacity of the vector for heterologous coding RNA. [0006]
  • We have also found it possible to provide a lentiviral vector based on a non-primate lentivirus which has a high coding capacity for heterologous coding sequences and which has a reduced capacity to transfer retroviral components to the target cell. [0007]
  • It has surprisingly been found that the amount of vector genomic sequence required from a non-primate lentivirus to produce an efficient cloning vector is substantially less than has been described for an HIV-based vector. [0008]
  • The sequence requirements for packaging HIV vector genomes are complex. The HIV-1 packaging signal encompasses the splice donor site and contains a portion of the 5′-untranslated region of the gag gene which has a putative secondary structure containing 4 short stem-loops. Additional sequences elsewhere in the genome are also known to be important for efficient encapsidation of HV. For example the first 350 bps of the gag protein coding sequence may contribute to efficient packaging and ill defined regions of env are also implicated. For the construction of HIV-vectors capable of expressing heterologous genes, a packaging signal extending to 350 bps of the gag protein-coding region has been used. We have surprisingly found that the structure of the packaging signal in non-primate lentiviruses is entirely different from that of HIV. Instead of a short sequence of 4 stem loops followed by an ill defined region of gag and env sequences, we have discovered that a shorter region of the gag gene suffices for efficient packaging. Indeed deletion of larger regions of the gag gene in EIAV vectors is advantageous and leads to higher titre viral vector being produced. This information can be used to provide improved vectors constructed from non-primate lentivirus sequences which have high titre and advantageous features compared to HV vectors. [0009]
  • In a first aspect of the invention, there is provided a retroviral vector genome containing a deleted gag gene from a non-primate lentivirus wherein the deletion in gag removes one or more nucleotides downstream of [0010] nucleotide 350 of the gag coding sequence. Preferably the deletion extends from nucleotide 350 to at least the C-terminus of the gagpol coding region. More preferably the deletion additionally removes nucleotide 300 of the gag coding region and most preferably the deletion retains only the first 150 nucleotides of the gag coding region. However even larger deletions of gag can also be used, for example the gag coding region contains the first 109 nucleotides of the gag coding region. It may also be possible for the gag coding region to contain only the first 2 nucleotides of the gag coding region.
  • Additional features of the lentiviral genome are included in the vector genome which are necessary for transduction of the target cell; replication; reverse transcription and integration. These are, at least, a portion of an LTR containing sequence from the R-region and U5 region, sequences from the 3′ LTR which contain a polypurine tract (PT) and a 3′ LTR from the non-primate lentivirus or a hybrid LTR containing sequences from the non-primate lentivirus and other elements. Optionally, the retroviral genome may contain accessory genes derived from a retrovirus, such as, but not limited to, a rev gene, a tat gene, a vif gene, a nef gene, a vpr gene or an S2 gene. Additional components may be added such as introns, splice-donor sites, a rev responsive element (RRE), cloning sites and selectable marker genes. [0011]
  • Moreover, we have now surprisingly demonstrated that a non-primate lentivirus minimal vector system can be constructed which requires neither S2, Tat, env nor dUTPase for either vector production or for transduction of dividing and non-dividing cells. [0012]
  • Thus according to another aspect the non-primate lentivirus genome from which the vector is derived lacks one or more accessory genes. [0013]
  • The deletion of accessory genes is highly advantageous. Firstly, it permits vectors to be produced without the genes normally associated with disease in lentiviral (e.g. HIV) infections. In particular, tat and env are associated with disease. Secondly, the deletion of accessory genes permits the vector to package more heterologous DNA. Thirdly, genes whose function is unknown, such as dUTPase and S2, may be omitted, thus reducing the risk of causing undesired effects. [0014]
  • In addition, we have shown that the leader sequence of the non-primate lentivirus genome is essential for high protein expression of gag and gagpol. [0015]
  • Therefore in a further aspect the non-primate lentivirus genome from which the vector is derived lacks the tat gene but includes the leader sequence between the end of the 5′ LTR and the ATG of gag. [0016]
  • These data further define a minimal essential set of functional components for an optimal lentiviral vector. A vector is provided with maximal genetic capacity and high titre, but without accessory genes that are either of unknown function (S2, UTPase), and therefore may present risk, or are analogues of HIV proteins that may be associated with AIDS (tat, env). [0017]
  • It will be appreciated that the present invention provides a retroviral vector derived from a non-primate lentivirus genome (1) comprising a deleted gag gene wherein the deletion in gag removes one or more nucleotides downstream of [0018] nucleotide 350 of the gag coding sequence; (2) wherein one or more accessory genes are absent from the non-primate lentivirus genome; (3) wherein the non-primate lentivirus genome lacks the tat gene but includes the leader sequence between the end of the 5ζ LTR and the ATG of gag; and combinations of (1), (2) and (3). In a preferred embodiment the retroviral vector comprises all of features (1) and (2) and (3).
  • A “non-primate” vector, as used herein, refers to a vector derived from a virus which does not primarily infect primates, especially humans. Thus, non-primate virus vectors include vectors which infect non-primate mammals, such as dogs, sheep and horses, reptiles, birds and insects. [0019]
  • A lentiviral or lentivirus vector, as used herein, is a vector which comprises at least one component part derived from a lentivirus. Preferably, that component part is involved in the biological mechanisms by which the vector infects cells, expresses genes or is replicated. [0020]
  • The non-primate lentivirus may be any member of the family of lentiviridae which does not naturally infect a primate and may include a feline immunodeficiency virus (F1v), a bovine immunodeficiency virus (BIV), a caprine arthritis encephalitis virus (CAEV), a Maedi visna virus (MVV) or an equine infectious anaemia virus (EIAV). Preferably the lentivirus is an EIAV. Equine infectious anaemia virus infects all equidae resulting in plasma viremia and thrombocytopenia (Clabough, et al. 1991. J Virol. 65:6242-51). Virus replication is thought to be controlled by the process of maturation of monocytes into macrophages. [0021]
  • EIAV has the simplest genomic structure of the lentiviruses. In addition to the gag, pol and env genes EIAV encodes three other genes: tat, rev, and S2. Tat acts as a transcriptional activator of the viral LTR (Derse and Newboldl993 Virology. 194:530-6; Maury, et al 1994 Virology. 200:632-42.) and Rev regulates and coordinates the expression of viral genes through rev-response elements (RRE) (Martarano et al 1994 J Virol. 68:3102-11.). The mechanisms of action of these two proteins are thought to be broadly similar to the analogous mechanisms in the primate viruses (Martano et al ibid). The function of S2 is unknown. In addition, an EIAV protein, Ttm, has been identified that is encoded by the first exon of tat spliced to the env coding sequence at the start of the transmembrane protein. [0022]
  • In addition to protease, reverse transcriptase and integrase non-primate lentiviruses contain a fourth pol gene product which codes for a dUTPase. This may play a role in the ability of these lentiviruses to infect certain non-dividing cell types. [0023]
  • The viral RNA in the first aspect of the invention is transcribed from a promoter, which may be of viral or non-viral origin, but which is capable of directing expression in a eukaryotic cell such as a mammalian cell. Optionally an enhancer is added, either upstream of the promoter or downstream. The RNA transcript is terminated at a polyadenylation site which may be the one provided in the lentiviral 3ζ LTR or a different polyadenylation signal. [0024]
  • Thus the present invention provides a DNA transcription unit comprising a promoter and optionally an enhancer capable of directing expression of a retroviral vector genome. [0025]
  • Transcription units as described herein comprise regions of nucleic acid containing sequences capable of being transcribed. Thus, sequences encoding MRNA, tRNA and rRNA are included within this definition. The sequences may be in the sense or antisense orientation with respect to the promoter. Antisense constructs can be used to inhibit the expression of a gene in a cell according to well-known techniques. Nucleic acids may be, for example, ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or analogues thereof. Sequences encoding mRNA will optionally include some or all of 5′ and/or 3′ transcribed but untranslated flanking sequences naturally, or otherwise, associated with the translated coding sequence. It may optionally further include the associated transcriptional control sequences normally associated with the transcribed sequences, for example transcriptional stop signals, polyadenylation sites and downstream enhancer elements. Nucleic acids may comprise cDNA or genomic DNA (which may contain introns). [0026]
  • The term “promoter” is used in the normal sense of the art, e.g. an RNA polymerase binding site in the Jacob-Monod theory of gene expression. [0027]
  • The term “enhancer” includes a DNA sequence which binds to other protein components of the transcription initiation complex and thus facilitates the initiation of transcription directed by its associated promoter. [0028]
  • The promoter and enhancer of the transcription units encoding the first viral vector component are preferably strongly active, or capable of being strongly induced, in the producer cell under conditions for production of the retroviral vector of the present invention and/or in primary target cells under conditions for production of the secondary viral vector. The promoter and enhancer of the transcription units encoding the second viral vector component are preferably strongly active, or capable of being strongly induced, in the target cells. The promoter and/or enhancer may be constitutively efficient, or may be tissue or temporally restricted in their activity. Examples of suitable tissue restricted promoters/enhancers are those which are highly active in tumour cells such as a promoter/enhancer from a MUC1 gene, a CEA gene or a 5T4 antigen gene. Examples of temporally restricted promoters/enhancers are those which are responsive to ischaemia and/or hypoxia, such as hypoxia response elements or the promoter/enhancer of a grp78 or a grp94 gene. One preferred promoter-enhancer combination is a human cytomegalovirus (hCMV) major immediate early (MIE) promoter/enhancer combination. [0029]
  • The LTRs may be altered in, for example, U3 (such as to obtain strong constitutive expression, inducible expression or tissue specific expression); R (such as to remove TAR stem loops); or U5 (such as to use enhanced non-U5 based polyadenylation signals, for example from the bovine growth hormone gene). [0030]
  • In one configuration the internal promoter cassette is reversed and a polyadenylation signal is placed downstream of the cassette. [0031]
  • In another embodiment the polyadenylation signal which is used contains at least one intron. [0032]
  • The vector of the present invention may make use of self-inactivating strategies. Self-inactivating retroviral vectors have been constructed by deleting the transcriptional enhancers or the enhancers and promoters in the U3 region of the 3′ LTR. After one round of vector replication, these changes are copied into both the 5′ and the 3′ LTRs producing an inactive provirus. However, any promoters internal to the LTRs in such vectors will still be active. This strategy has been employed to eliminate effects of the enhancers and promoters in the viral LTRs on transcription from internally placed genes. Such effects include increased transcription or suppression of transcription. This strategy can also be used to eliminate downstream transcription from the 3′ LTR into genomic DNA. This is of particular concern in human gene therapy where it is of critical importance to prevent any activation of an endogenous oncogene. [0033]
  • Another type of self-inactivating vector has been constructed that has direct repeats flanking the packaging signal such that the packaging signal is frequently deleted during reverse transcription, producing virus defective for packaging. With sufficiently long direct repeats, a majority of resultant proviruses lose their packaging sequences. The rate of deletion could be increased to 100% by designing the vector so that packaging signal deletion reconstituted the neo marker nad be selecting the vector-infected cells in G418. This strategy may be particularly useful for gene therapy applications where any spread of the vector following gene transfer is undesirable. [0034]
  • In a further preferred embodiment of the first aspect of the invention, one or more nucleotides of interest (NOI) is introduced into the vector at the cloning site. Such therapeutic genes may be expressed from a promoter placed in the retroviral LTR or may be expressed from an internal promoter introduced at the cloning site. [0035]
  • Suitable NOI coding sequences include those that are of therapeutic and/or diagnostic application such as, but are not limited to: sequences encoding cytokines, chemokines, hormones, antibodies, engineered immunoglobulin-like molecules, a single chain antibody, fusion proteins, enzymes, immune co-stimulatory molecules, immunomodulatory molecules, anti-sense RNA, a transdominant negative mutant of a target protein, a toxin, a conditional toxin, an antigen, a tumour suppressor protein and growth factors, membrane proteins, vasoactive proteins and peptides, anti-viral proteins and ribozymes, and derivatives therof (such as with an associated reporter group). When included, such coding sequences may be typically operatively linked to a suitable promoter, which may be a promoter driving expression of a ribozyme(s), or a different promoter or promoters. [0036]
  • The NOI coding sequence may encode a fusion protein or a segment of a coding sequence. [0037]
  • The retroviral vector of the present invention may be used to deliver a NOI such as a pro-drug activating enzyme to a tumour site for the treatment of a cancer. In each case, a suitable pro-drug is used in the treatment of the individual (such as a patient) in combination with the appropriate pro-drug activating enzyme. An appropriate pro-drug is administered in conjunction with the vector. Examples of pro-drugs include: etoposide phosphate (with alkaline phosphatase, Senter et al 1988 Proc Natl Acad Sci 85: 4842-4846); 5-fluorocytosine (with cytosine deaminase, Mullen et al 1994 Cancer Res 54: 1503-1506); Doxorubicin-N-p-hydroxyphenoxyacetamide (with Penicillin-V-Amidase, Kerr et al 1990 Cancer Immunol Immunother 31: 202-206); Para-N-bis(2-chloroethyl) aminobenzoyl glutamate (with carboxypeptidase G2); Cephalosporin nitrogen mustard carbamates (with β-lactamase); SR4233 (with P450 Reducase); Ganciclovir (with HSV thymidine kinase, Borrelli et al 1988 Proc Natl Acad Sci 85: 7572-7576); mustard pro-drugs with nitroreductase (Friedlos et al 1997 J Med Chem 40: 1270-1275) and Cyclophosphamide (with P450 Chen et al 1996 Cancer Res 56: 1331-1340). [0038]
  • The vector of the present invention may be delivered to a target site by a viral or a non-viral vector. [0039]
  • As it is well known in the art, a vector is a tool that allows or faciliates the transfer of an entity from one environment to another. By way of example, some vectors used in recombinant DNA techniques allow entities, such as a segment of DNA (such as a heterologous DNA segment, such as a heterologous cDNA segment), to be transferred into a target cell. Optionally, once within the target cell, the vector may then serve to maintain the heterologous DNA within the cell or may act as a unit of DNA replication. Examples of vectors used in recombinant DNA techniques include plasmids, chromosomes, artificial chromosomes or viruses. [0040]
  • Non-viral delivery systems include but are not limted to DNA transfection methods. Here, transfection includes a process using a non-viral vector to deliver a gene to a target mammalian cell. [0041]
  • Typical transfection methods include electroporation, DNA biolistics, lipid-mediated transfection, compacted DNA-mediated transfection, liposomes, immunoliposomes, lipofectin, cationic agent-mediated, cationic facial amphiphiles (CFAs) (Nature Biotechnology 1996 14; 556), and combinations thereof. [0042]
  • Viral delivery systems include but are not limited to adenovirus vector, an adeno-associated viral (AAV) vector, a herpes viral vector, retroviral vector, lentiviral vector, baculoviral vector. Other examples of vectors include ex vivo delivery systems, which include but are not limited to DNA transfection methods such as electroporation, DNA biolistics, lipid-mediated transfection, compacted DNA-mediated transfection. [0043]
  • The term “retroviral vector particle” refers to the packaged retroviral vector, that is preferably capable of binding to and entering target cells. The components of the particle, as already discussed for the vector, may be modified with respect to the wild type retrovirus. For example, the Env proteins in the proteinaceous coat of the particle may be genetically modified in order to alter their targeting specificity or achieve some other desired function. [0044]
  • Preferably, the viral vector preferentially transduces a certain cell type or cell types. [0045]
  • More preferably, the viral vector is a targeted vector, that is it has a tissue tropism which is altered compared to the native virus, so that the vector is targeted to particular cells. [0046]
  • For retroviral vectors, this may be achieved by modifying the Env protein. The Env protein of the retroviral secondary vector needs to be a non-toxic envelope or an envelope which may be produced in non-toxic amounts within the primary target cell, such as for example a MMLV amphotropic envelope or a modified amphotropic envelope. The safety feature in such a case is preferably the deletion of regions or sequence homology between retroviral components. [0047]
  • Preferably the envelope is one which allows transduction of human cells. Examples of suitable env genes include, but are not limited to, VSV-G, a MLV amphotropic env such as the 4070A env, the-RD114 feline leukaemia virus env or haemagglutinin (HA) from an influenza virus. The Env protein may be one which is capable of binding to a receptor on a limited number of human cell types and may be an engineered envelope containing targeting moieties. The env and gag-pol coding sequences are transcribed from a promoter and optionally an enhancer active in the chosen packaging cell line and the transcription unit is terminated by a polyadenylation signal. For example, if the packaging cell is a human cell, a suitable promoter-enhancer combination is that from the human cytomegalovirus major immediate early (hCMV-MIE) gene and a polyadenylation signal from SV40 virus may be used. Other suitable promoters and polyadenylation signals are known in the art. [0048]
  • The packaging cell may be an in vivo packaging cell in the body of an individual to be treated-orit may be a cell cultured in vitro such as a tissue culture cell line. Suitable cell lines include mammalian cells such as murine fibroblast derived cell lines or human cell lines. Preferably the packaging cell line is a human cell line, such as for example: 293 cell line, HEK293, 293-T, TE671, HT1080. [0049]
  • Alternatively, the packaging cell may be a cell derived from the individual to be treated such as a monocyte, macrophage, stem cells, blood cell or fibroblast. The cell may be isolated from an individual and the packaging and vector components administered ex vivo followed by re-administration of the autologous packaging cells. Alternatively the packaging and vector components may be administered to the packaging cell in vivo. Methods for introducing retroviral packaging and vector components into cells of an individual are known in the art. For example, one approach is to introduce the different DNA sequences that are required to produce a retroviral vector particle e.g. the env coding sequence, the gag-pol coding sequence and the defective retroviral genome into the cell simultaneously by transient triple transfection (Landau & Littman 1992 J. Virol. 66, 5110; Soneoka et al 1995 Nucleic Acids Res 23:628-633). [0050]
  • In one embodiment the vector configurations of the present invention use as their production system, three transcription units expressing a genome, the gag-pol components and an envelope. The envelope expression cassette may include one of a number of envelopes such as VSV-G or various murine retrovirus envelopes such as 4070A. [0051]
  • Conventionally these three cassettes would be expressed from three plasmids transiently transfected into an appropriate cell line such as 293T or from integrated copies in a stable producer cell line. An alternative approach is to use another virus as an expression system for the three cassettes, for example baculovirus or adenovirus. These are both nuclear expression systems. To date the use of a poxvirus to express all of the components of a retroviral or lentiviral vector system has not been described. In particular, given the unusual codon usage of lentiviruses and their requirement for RNA handling systems such as the rev/RRE system it has not been clear whether incorporation of all three cassettes and their subsequent expression in a vector that expresses in the cytoplasm rather than the nucleus is feasible. Until now the possibility remained that key nuclear factors and nuclear RNA handling pathways would be required for expression of the vector components and their function in the gene delivery vehicle. Here we describe such a system and show that lentiviral components can be made in the cytoplasm and that they assemble into functional gene delivery systems. The advantage of this system is the ease with which poxviruses can be handled, the high expression levels and the ability to retain introns in the vector genomes. [0052]
  • According to another aspect therefore there is provided a hybrid viral vector system for in vivo gene delivery, which system comprises a primary viral vector which is obtainable from or is based on a poxvirus and a second viral vector which is obtainable from or is based on a vectroviral vector, preferably a lentiviral vector, even more preferably a non-primate lentiviral vector. [0053]
  • The secondary vector may be produced from expression of essential genes for retroviral vector production encoded in the DNA of the primary vector. Such genes may include a gag-pol from a retrovirus, an env gene from an enveloped virus and a defective retroviral vector containing one or more therapeutic or diagnostic NOI(s). The defective retroviral vector contains in general terms sequences to enable reverse transcription, at least part of a 5′ long terminal repeat (LTR), at least part of a 3′LTR and a packaging signal. [0054]
  • If it is desired to render the secondary vector replication defective, that secondary vector may be encoded by a plurality of transcription units, which may be located in a single or in two or more adenoviral or other primary vectors. [0055]
  • In some therapeutic or experimental situations it may be desirable to obviate the need to make EAIV derived from MVA in vitro. MVA-EIAV hybrids are delivered directly into the patient/animal e.g. MVA-EIAV is injected intravenously into the tail vein of a mouse and this recombinant virus infects a variety of murine tissues e.g. lung, spleen etc. Infected cells express transduction competent EIAV containing a therapeutic gene for gene therapy for example. EIAV vector particles bud from these cells and transduce neighbouring cells. The transduced cell then contains an integrated copy of the EIAV vector genome and expresses the therapeutic gene product or other gene product of interest. If expression of the therapeutic gene product is potentially toxic to the host it may be regulated by a specific promoter, e.g. the hypoxic response element (HRE), which will restrict expression to those cells in a hypoxic environment. For gene therapy of lung/trachea epithelium cells e.g to treat cystic fibrosis MVA-EIAV may be given as an aerosol delivered intranasally. Alternatively, macrophages can be transduced in vitro and then reintroduced to create macrophage factories for EIAV-based vectors. Furthermore, because MVA is replication incompetent MVA-EIAV hybrids could also be used to treat immuno-suppressed hosts. [0056]
  • Vaccinia virus, the prototypic member of the orthopox genus within the family poxviridae, was the first virus used for expression of recombinant exogenous proteins (Mackett et al 1982, Paoletti & Panicalli 1982). Vaccinia virus has a large DNA genome of greater than 180 kb and reports indicate that it can accommodate over 25 kb of foreign DNA (Merchlinsky & Moss 1992). Several other strains of poxviruses have been adapted as recombinant expression vectors (for review see Carroll and Moss 1997) e.g. fowlpox (Taylor & Paoletti 1988), canarypox (Taylor et al 1991), swinepox (van der Leek et al 1994) and entomopox (Li et al 1997). Additionally, due to safety concerns, several highly attenuated strains of vaccinia virus have been developed that are compromised in human and other mammalian cells e.g. modified vaccinia virus Ankara (MVA) (Mayr 1978, Sutter 1992), NYVAC (Paoletti et al 1994), vaccinia virus deficient in a DNA replication enzyme (Holzer et al 1997). These may all be used in the present invention. [0057]
  • MVA was derived from a replication competent vaccinia smallpox vaccine strain, Ankara. After >500 passages in chick embryo fibroblast cells the virus isolate was shown to be highly attenuated in a number of animal models including mice that were immune deficient (Mayr et al 1978). The attenuated isolate, MVA, was used to vaccinate over 120,000 people, many of which were immunocompromised (Mahnel 1994) without adverse effects. Studies illustrate that MVA can infect a wide range of mammalian cells but productive infection has only been observed in Hamster kidney cell BHK-21 (Carroll 1997). In all other tested mammalian cell lines early expression, DNA replication and late expression are observed leading to the production of non-infectious immature virus particles (Carroll 1997, Meyer 1991). Virus replication studies show that a minority of mammalian cells can support very low level production of infectious virus i.e. BS-C-1 cells in which 1 infectious MVA particle is produced per cell (Carroll and Moss 1997). Late gene expression usually give rise to >10 fold more protein that those genes under early promoters (Chakrabarti et al 1997, Wyatt et al 1996). In all other attenuated poxvirus strains late gene expression is rarely observed in mammalian cells. [0058]
  • Production of retrovirus vector systems e.g. MLV-HIV and lentivirus vector systems requires the construction of producer lines that express the virus genome and essential structural proteins to make transduction competent virus. Generally, this is a relatively inefficient process which is further complicated when the virus is pseudotyped with toxic envelope proteins such as VSV-G. Expression of a functional genome and the required structural proteins from within a recombinant poxvirus may obviate many of the current inefficient retrovirus and lentivirus vector production technologies. Additionally, such recombinant poxviruses may be directly injected into patients to give rise to in vivo production of retrovirus or lentivirus. [0059]
  • MVA is a particularly suitable poxvirus for the construction of a pox-retrovirus or pox-lentivirus hybrid due to its non-replicating phenotype and its ability to perform both early and strong late expression for the production of high titre vector preparations. [0060]
  • In order to produce a functional retrovirus or lentivirus vector genome it is essential that the 5′ of the RNA genome should be exact (Cannon et al. 1996). This is a challenge in a vaccinia-based production system as many of the vaccinia promoter comprise downstream determinants of transcription efficiency (Davison 1989b, Moss 1996). However, we show that thereare several ways to solve this problem: [0061]
  • a. Use of a T7 promoter and T7 termination sequence. [0062]
  • b. Use of early promoters (in which sequences downstream of the RNA start site are not highly conserved), (Davison 1989a). [0063]
  • c. Use of intermediate and late promoters of vaccinia which require additional sequences downstream of the initiation site in conjunction with strategies to generate an authentic 5′ end or which place the additional downstream sequences into both R regions. There is a requirement for specific sequences of 4 nucleotides downstream of the initiation of transcription in the late promoter (Davison 1989b, Moss 1996). In the first case a ribozyme is placed downstream of the promoter and upstream of the R region. The ribozyme is designed to cleave the RNA in cis to generate the correct 5′ end. In the second the approach is to modify the R regions to incorporate the extra sequences. This must be done in both the 5′ and the 3′ LTR R regions. [0064]
  • The advantage of having a T7 dependent system is that it would require the infection of the cell by two recombinant vaccinia viruses to produce transducing EIAV viral particles. For example, one MVA could carry the vector genome, under the control of the T7 promoter and the gag/pol and the env sequences under the control of the vaccinia promoters. The other MVA would carry the T7 polymerase gene under the control of a vaccinia promoter (Wyatt et al 1995). [0065]
  • The retroviral vector particle according to the invention will also be capable of transducing cells which are slowly-dividing, and which non-lentiviruses such as MLV would not be able to efficiently transduce. Slowly-dividing cells divide once in about every three to four days including certain tumour cells. Although tumours contain rapidly dividing cells, some tumour cells especially those in the centre of the tumour, divide infrequently. Alternatively the target cell may be a growth-arrested cell capable of undergoing cell division such as a cell in a central portion of a tumour mass or a stem cell such as a haematopoietic stem cell or a CD34-positive cell. As a further alternative, the target cell may be a precursor of a differentiated cell such as a monocyte precursor, a CD33-positive cell, or a myeloid precursor. As a further alternative, the target cell may be a differentiated cell such as a neuron, astrocyte, glial cell, microglial cell, macrophage, monocyte, epithelial cell, endothelial cell or hepatocyte. Target cells may be transduced either in vitro after isolation from a human individual or may be transduced directly in vivo. [0066]
  • The delivery of one or more therapeutic genes by a vector system according to the present invention may be used alone or in combination with other treatments or components of the treatment. [0067]
  • For example, the retroviral vector of the present invention may be used to deliver one or more NOI(s) useful in the treatment of the disorders listed in WO-A-98/05635. For ease of reference, part of that list is now provided: cancer, inflammation or inflammatory disease, dermatological disorders, fever, cardiovascular effects, haemorrhage, coagulation and acute phase response, cachexia, anorexia, acute infection, HIV infection, shock states, graft-versus-host reactions, autoimmune disease, reperfusion injury, meningitis, migraine and aspirin-dependent anti-thrombosis; tumour growth, invasion and spread, angiogenesis, metastases, malignant, ascites and malignant pleural effusion; cerebral ischaemia, ischaemic heart disease, osteoarthritis, rheumatoid arthritis, osteoporosis, asthma, multiple sclerosis, neurodegeneration, Alzheimer's disease, atherosclerosis, stroke, vasculitis, Crohn's disease and ulcerative colitis; periodontitis, gingivitis; psoriasis, atopic dermatitis, chronic ulcers, epidermolysis bullosa; corneal ulceration, retinopathy and surgical wound healing; rhinitis, allergic conjunctivitis, eczema, anaphylaxis; restenosis, congestive heart failure, endometriosis, atherosclerosis or endosclerosis. [0068]
  • In addition, or in the alternative, the retroviral vector of the present invention may be used to deliver one or more NOI(s) useful in the treatment of disorders listed in WO-A-98/07859. For ease of reference, part of that list is now provided: cytokine and cell proliferation/differentiation activity; immunosuppressant or immunostimulant activity (e.g. for treating immune deficiency, including infection with human immune deficiency virus; regulation of lymphocyte growth; treating cancer and many autoimmune diseases, and to prevent transplant rejection or induce tumour immunity); regulation of haematopoiesis, e.g. treatment of myeloid or lymphoid diseases; promoting growth of bone, cartilage, tendon, ligament and nerve tissue, e.g. for healing wounds, treatment of burns, ulcers and periodontal disease and neurodegeneration; inhibition or activation of follicle-stimulating hormone (modulation of fertility); chemotactic/chemokinetic activity (e.g. for mobilising specific cell types to sites of injury or infection); haemostatic and thrombolytic activity (e.g. for treating haemophilia and stroke); antiinflammatory activity (for treating e.g. septic shock or Crohn's disease); as antimicrobials; modulators of e.g. metabolism or behaviour; as analgesics; treating specific deficiency disorders; in treatment of e.g. psoriasis, in human or veterinary medicine. [0069]
  • In addition, or in the alternative, the retroviral vector of the present invention may be used to deliver one or more NOI(s) useful in the treatment of disorders listed in WO-A-98/09985. For ease of reference, part of that list is now provided: macrophage inhibitory and/or T cell inhibitory activity and thus, anti-inflammatory activity; anti-immune activity, i.e. inhibitory effects against a cellular and/or humoral immune response, including a response not associated with inflammation; inhibit the ability of macrophages and T cells to adhere to extracellular matrix components and fibronectin, as well as up-regulated fas receptor expression in T cells; inhibit unwanted immune reaction and inflammation including arthritis, including rheumatoid arthritis, inflammation associated with hypersensitivity, allergic reactions, asthma, systemic lupus erythematosus, collagen diseases and other autoimmune diseases, inflammation associated with atherosclerosis, arteriosclerosis, atherosclerotic heart disease, reperfusion injury, cardiac arrest, myocardial infarction, vascular inflammatory disorders, respiratory distress syndrome or other cardiopulmonary diseases, inflammation associated with peptic ulcer, ulcerative colitis and other diseases of the gastrointestinal tract, hepatic fibrosis, liver cirrhosis or other hepatic diseases, thyroiditis or other glandular diseases, glomerulonephritis or other renal and urologic diseases, otitis or other oto-rhino-laryngological diseases, dermatitis or other dermal diseases, periodontal diseases or other dental diseases, orchitis or epididimo-orchitis, infertility, orchidal trauma or other immune-related testicular diseases, placental dysfunction, placental insufficiency, habitual abortion, eclampsia, pre-eclampsia and other immune and/or inflammatory-related gynaecological diseases, posterior uveitis, intermediate uveitis, anterior uveitis, conjunctivitis, chorioretinitis, uveoretinitis, optic neuritis, intraocular inflammation, e.g. retinitis or cystoid macular oedema, sympathetic ophthalmia, scleritis, retinitis pigmentosa, immune and inflammatory components of degenerative fondus disease, inflammatory components of ocular trauma, ocular inflammation caused by infection, proliferative vitreo-retinopathies, acute ischaemic optic neuropathy, excessive scarring, e.g. following glaucoma filtration operation, immune and/or inflammation reaction against ocular implants and other immune and inflammatory-related ophthalmic diseases, inflammation associated with autoimmune diseases or conditions or disorders where, both in the central nervous system (CNS) or in any other organ, immune and/or inflammation suppression would be beneficial, Parkinson's disease, complication and/or side effects from treatment of Parkinson's disease, AIDS-related dementia complex HIV-related encephalopathy, Devic's disease, Sydenham chorea, Alzheimer's disease and other degenerative diseases, conditions or disorders of the CNS, inflammatory components of stokes, post-polio syndrome, immune and inflammatory components of psychiatric disorders, myelitis, encephalitis, subacute sclerosing pan-encephalitis, encephalomyelitis, acute neuropathy, subacute neuropathy, chronic neuropathy, Guillaim-Barre syndrome, Sydenham chora, myasthenia gravis, pseudo-tumour cerebri, Down's Syndrome, Huntington's disease, amyotrophic lateral sclerosis, inflammatory components of CNS compression or CNS trauma or infections of the CNS, inflammatory components of muscular atrophies and dystrophies, and immune and inflammatory related diseases, conditions or disorders of the central and peripheral nervous systems, post-traumatic inflammation, septic shock, infectious diseases, inflammatory complications or side effects of surgery, bone marrow transplantation or other transplantation complications and/or side effects, inflammatory and/or immune complications and side effects of gene therapy, e.g. due to infection with a viral carrier, or inflammation associated with AIDS, to suppress or inhibit a humoral and/or cellular immune response, to treat or ameliorate monocyte or leukocyte proliferative diseases, e.g. leukaemia, by reducing the amount of monocytes or lymphocytes, for the prevention and/or treatment of graft rejection in cases of transplantation of natural or artificial cells, tissue and organs such as cornea, bone marrow, organs, lenses, pacemakers, natural or artificial skin tissue. [0070]
  • The present invention also provides a pharmaceutical composition for treating an individual by gene therapy, wherein the composition comprises a therapeutically effective amount of the retroviral vector of the present invention comprising one or more deliverable therapeutic and/or diagnostic NOI(s) or a viral particle produced by or obtained from same. The pharmaceutical composition may be for human or animal usage. Typically, a physician will determine the actual dosage which will be most suitable for an individual subject and it will vary with the age, weight and response of the particular individual. [0071]
  • The composition may optionally comprise a pharmaceutically acceptable carrier, diluent, excipient or adjuvant. The choice of pharmaceutical carrier, excipient or diluent can be selected with regard to the intended route of administration and standard pharmaceutical practice. The pharmaceutical compositions may comprise as—or in addition to—the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s), and other carrier agents that may aid or increase the viral entry into the target site (such as for example a lipid delivery system). [0072]
  • Where appropriate, the pharmaceutical compositions can be administered by any one or more of: inhalation, in the form of a suppository or pessary, topically in the form of a lotion, solution, cream, ointment or dusting powder, by use of a skin patch, orally in the form of tablets containing excipients such as starch or lactose, or in capsules or ovules either alone or in admixture with excipients, or in the form of elixirs, solutions or suspensions containing flavouring or colouring agents, or they can be injected parenterally, for example intracavemosally, intravenously, intramuscularly or subcutaneously. For parenteral administration, the compositions may be best used in the form of a sterile aqueous solution which may contain other substances, for example enough salts or monosaccharides to make the solution isotonic with blood. For buccal or sublingual administration the compositions may be administered in the form of tablets or lozenges which can be formulated in a conventional manner. [0073]
  • The delivery of one or more therapeutic genes by a vector system according to the invention may be used alone or in combination with other treatments or components of the treatment. Diseases which may be treated include, but are not limited to: cancer, neurological diseases, inherited diseases, heart disease, stroke, arthritis, viral infections and diseases of the immune system. Suitable therapeutic genes include those coding for tumour suppressor proteins, enzymes, pro-drug activating enzymes, immunomodulatory molecules, antibodies, engineered immunoglobulin-like molecules, fusion proteins, hormones, membrane proteins, vasoactive proteins or peptides, cytokines, chemokines, anti-viral proteins, antisense RNA and ribozymes. [0074]
  • In a preferred embodiment of a method of treatment according to the invention, a gene encoding a pro-drug activating enzyme is delivered to a tumour using the vector system of the invention and the individual is subsequently treated with an appropriate pro-drug. Examples of pro-drugs include etoposide phosphate (used with alkaline phosphatase Senter et al., 1988 Proc. Natl. Acad. Sci. 85: 4842-4846); 5-fluorocytosine (with Cytosine deaminase Mullen et al. 1994 Cancer Res. 54: 1503-1506); Doxorubicin-N-p-hydroxyphenoxyacetamide (with Penicillin-V-Amidase (Kerr et al. 1990 Cancer Immunol. Immunother. 31: 202-206); Para-N-bis(2-chloroethyl) aminobenzoyl glutamate (with Carboxypeptidase G2); Cephalosporin nitrogen mustard carbamates (with b-lactamase); SR4233 (with P450 Reducase); Ganciclovir (with HSV thymidine kinase, Borrelli et al. 1988 Proc. Natl. Acad. Sci. 85: 7572-7576) mustard pro-drugs with nitroreductase (Friedlos et al. 1997J Med Chem 40: 1270-1275) and Cyclophosphamide or Ifosfamide (with a cytochrome P450 Chen et al. 1996 Cancer Res 56: 1331-1340). [0075]
  • In accordance with the invention, standard molecular biology techniques may be used which are within the level of skill in the art. Such techniques are fully described in the literature. See for example; Sambrook et al (1989) Molecular Cloning; a laboratory manual; Hames and Glover (1985-1997) DNA Cloning: a practical approach, Volumes I-IV (second edition); Methods for the engineering of immunoglobulin genes are given in McCafferty et al (1996) “Antibody Engineering: A Practical Approach”. [0076]
  • The invention will now be further described by way of example in which reference is made to the following Figures in which: [0077]
  • FIG. 1—shows the structure of transcription units from plasmids pESP, pONY3 and pONY2.lnlsLacZ. [0078]
  • FIG. 2—shows a PCR analysis of integrated EIAV vector. PCR was performed with either genomic DNA from EIAV vector transduced cells ([0079] lanes 1 and 5) or mock transduced cells (lanes 2 and 6). pONY2.lnlsLacZ (lanes 3 and 7) and pONY3 (lanes 4 and 8) were used as controls. A. PCR detection of EIAV LTRs. B. PCR detection of pol.
  • FIG. 3—shows the structure of vector transcription units in deletion plasmids used to identify the packaging requirements for an EIAV vector. [0080]
  • FIG. 4—shows a secondary structure prediction for the RNA derived from the gag-transcription unit present in pONY2.13LacZ. [0081]
  • FIG. 5 is a representation of vectors derived from the EIAV genome. [0082]
  • FIG. 6 is a representation of gagpol constructs derived from EIAV. [0083]
  • FIG. 7 is a representation of an EIAV vector comprising an S2 deletion [0084]
  • FIG. 8 is a representation of EIAV gagpol constructs having deleted S2 and dUTPase genes. [0085]
  • FIG. 9 is a representation of an EIAV minimal vector. [0086]
  • FIG. 10 illustrates a gel showing an analysis of EIAV gagpol constructs according to the invention. [0087]
  • FIG. 11 shows examples of the pONY4 vectors. [0088]
  • FIG. 12 shows two SIN vectors. [0089]
  • FIG. 13 is a representation of a vector with a split polyA signal. [0090]
  • FIG. 14 is a representation of a vector with a split polyA signal. [0091]
  • FIG. 15 is a representation of a vector with a split polyA signal. [0092]
  • FIG. 16 shows construction of pONY4-GFP with a split polyA signal. [0093]
  • FIG. 17 shows construction of a MLV/EIAV vector. [0094]
  • FIG. 18 shows primers for construction of MLV/EIAV vectors. [0095]
  • FIG. 19 shows complete sequence of pONY-mouse. [0096]
  • FIGS. 20 and 21 give PCR priming. [0097]
  • FIG. 22 shows pEMVA4 (after PCR with primers EMVA 1-8). [0098]
  • FIG. 23 shows pEMVA4. [0099]
  • FIG. 24 shows pEMVA5. [0100]
  • FIGS. 25 and 26 show an example of hammer-head strategy for 5′ end formation. [0101]
  • FIG. 27 shows pEMVA6. [0102]
  • FIG. 28 shows pEMVA7 and pSyn pONY4.1. [0103]
  • FIG. 29 shows EMVA 10/11. [0104]
  • FIG. 30 shows pEMVA9. [0105]
  • FIG. 31 shows pEMVA10. [0106]
  • FIG. 32 shows pLWHORSE3.1. [0107]
  • FIG. 33 shows pMCRev. [0108]
  • FIG. 34 shows pYFVSVG. [0109]
  • FIG. 35 shows pYFAmpho. [0110]
  • FIG. 36 shows recombinant MVA constructs. [0111]
  • FIG. 37 shows the complete sequence of pSC65. [0112]
  • FIG. 38 shows the complete sequence of pLW22[0113]
  • In more detail, FIG. 1. Plasmids used in this study. The genomic organization of EIAV is indicated including splice donor (d1, d2 and d3) and splice acceptor sites (a1, a2 and a3). The positions of gag, pol, env, tat, rev, S2 and the viral LTRs are also shown. Plasmid pESP is an EIAV vector genome containing the SV40 promoter and the puromycin resistance gene. Plasmid pONY3 is an EIAV gagpol expression plasmid. pONY2.lnlslacZ is an EIAV vector genome containing a HCMV IE enhancer/promoter and β-galactosidase gene (nlslacZ). [0114]
  • FIG. 2 shows PCR analysis of integrated EIAV vector. PCR was performed with either genomic DNA from EIAV vector transduced cells ([0115] lanes 1 and 5) or mock transduced cells (lanes 2 and 6). pONY2.lnlsLacZ (lanes 3 and 7) and pONY3 (lanes 4 and 8) were used as controls. A. PCR detection of EIAV LTRs. B. PCR detection of pol.
  • TABLE 2. Transduction of dividing and non-dividing cells. 293T cells were treated with (open columns) or without (shaded columns) aphidicolin according to the method of Lewis et al. (Lewis, P. F., and M. Emerman. 1994. J Virol. 68:510-6.) and then transduced with either EIAV vector PONY2.lnlsLacZ or MLV vector HIT111 (Soneoka et al 1995 Nucl. Acids Res. 23:628-633). 48 hours later the cells were stained with X-gal. Titers were averaged from two independent experiments and calculated as lac Z colony forming units per ml. There was no more than 10% variation between experiments. [0116]
  • FIG. 10 shows analysis of gagpol expression constructs. 30%lg of total cellular protein was separated by SDS/Page electrophoresis, transferred to nitro-cellulose and probed with anti-EIAV antibodies. The secondary antibody was anti-Horse HRP (Sigma). Titres were averaged from three independent experiments and calulated as lacZ forming units per ml. There was no more than 10% variation between experiments. pONY2.lnlslacZ and the envelope expression plasmids were co-transfected with the EIAV gagpol. [0117]
  • EXAMPLE 1 Construction of EIAV Vectors Containing Deleted Gag Genes
  • In order to construct a replication incompetent EIAV vector system we have used, as a starting point, an infectious proviral clone pSPEIAV19 (accession number: U01866), described by Payne et al. (1994, J Gen Virol. 75:425-9). An initial EIAV based vector was constructed by simply deleting part of env by removing a Hind III/Hind III fragment corresponding to coordinates 5835/6571 according to the numbering system of Payne et al. (ibid.). This fragment was replaced with the puromycin resistance gene under the control of the SV40 early promoter from pTIN500 (Cannon et al 1996 J. Virol. 70:8234-8240) to create pESP (FIG. 1). Viral stocks were produced by calcium phosphate transfection of 293T cells (Soneoka et al 1995 Nucl. Acids Res. 23:628-633) with pESP and pRV67 (Kim et al 1998 J. Virol. 72(1):811-6) a plasmid in which the vesicular stomatitis virus glycoprotein (VSV-G) is expressed from the HCMV-IE enhancer/promoter. Alternatively other VSV-G expression plasmids can be used eg Yee, et al 1994 PNAS 91:9564-9568. The resulting supernatant was used to transduce human kidney (293T) and canine osteosarcoma cells (D17) as follows. 48 hours post-transfection tissue culture fluid was collected and filtered through 0.45 cm filters. Ten-fold dilutions were made in culture medium containing polybrene at 8 μg/ml and then 500 μl aliquots placed on D17 cells seeded at 1.6×10[0118] 5/well in 12 well plates on the previous day. Two hours later 1 ml of culture media was added. Two days later puromycin was added to a final concentration of 4 ug/ml and incubation was continued for a further 7 days. As a positive control, a Murine leukaemia virus (MLV) based vector (pTIN500) containing the puromycin resistance gene under the control of the SV40 early promoter was used in conjunction with pHIT60 (MLV gagpol) and pRV67 (Cannon et al 1996 J. Virol. 70:8234-8240). No resistance colonies were detected on either cell type after 7 days of puromycin selection with the EIAV vector. The MLV vector produced 5.0×104 c.f.u./ml on 293T cells and 1.0×104 c.f.u./ml on D17 cells.
  • The likely explanation for this result is that the EIAV LTR is not functional in human cells in the absence of tat and so insufficient amounts of the critical components such as gag-pol and tat are produced. [0119]
  • A further vector system was therefore constructed comprising three transcription units to produce the following: 1) vector genome RNA; 2) env and 3) gag-pol. In order to ensure that sufficient of each component is produced, the env and gag-pol transcription units are transcribed from a promoter-enhancer active in the chosen human packaging cell line. In this way, sufficient gag-pol and, most likely tat, are produced to ensure efficient production of transduction-competent vector particles. [0120]
  • The vector genome was constructed which has the reporter gene within the pol region of the genome as follows. The plasmid designated pONYl was constructed by inserting the EIAV LTR, amplified by PCR from pSPEIAV19, into pBluescript II KS+(Stratagene). The 5′ LTR of EIAV clone pSPEIAV19 was PCR amplified using pfu polymerase with the following primers: [0121]
  • 5′ GCATGGACCTGTGGGGTTTTTATGAGG [0122]
  • 3′ GCATGAGCTCTGTAGGATCTCGAACAGAC [0123]
  • The amplicon was blunt ended by 5′ overhang fill-in and inserted into pBluescript II KS+cut with Bss HII which had been blunt ended by 3′ overhand removal using T4 DNA polymerase. This construct was called pONY1 and the orientation was 5′ to 3′ in relation to β-galactosidase of pBluescript II KS+. Sequencing of pONY1 revealed no mutations. [0124]
  • Vector genome pSPEIAV19DH was cut with Mlu I (216/8124) and inserted into pONY1 Mlu I cut (216) to make pONY2. A Bss HII digest (619/792) of pBluescript II KS+ was carried out to obtain the multiple cloning site. This was blunt ended by 5′ overhang fill-in and ligated to pONY2 cut with Bgl II and Nco I (1901/4949) and blunt ended by 5′ overhang fill-in. The orientation was 3′ to 5′ in relation to the EIAV sequence. This was called pONY2.1. pSPCMV was created by inserting pLNCX (Accession number: M28246) (Pst I/Hind III) into pSP72 (Promega). The β-galactosidase gene was inserted from pTIN414 (Cannon PM et al J. Virol. 70, 8234-8240) into pSP72 (Xho I/Sph I) to make pSPlacZ. The 5′ end to the β-galactosidase gene was replaced by the SV40 T antigen nuclear localization signal from pAD.RSVbgal (J. Clin. Invet. 90:626-630, 1992). pAD.RSVbgal was cut with Xho I/Cla I and inserted into Xho I/Cla I pSPlacZ to make pSPnlslacZ. The CMV nuclear localizing and non nuclear localizing β-galactosidase from pSPlacZ and pSPnlslacZ was cut out with Pst I and inserted into the Pst I site of pONY2.1 in the 5′ to 3′ orientation of EIAV. These were called pONY2. lnlslacZ and pONY2.1lacZ. [0125]
  • An EIAV gagpol expression plasmid (pONY3) was then made by inserting Mlu I/Mlu I fragment from pONY[0126] 2ΔH into the mammalian expression plasmid pCl-neo (Promega) such that the gag-pol gene is expressed from the hCMV-MIE promoter-enhancer. In particular, gagpol pSPEIAV19DH was cut with Mlu I (216/8124) and inserted into pCI-Neo (Promega) Mlu I cut (216) to make pONY3. Plasmid pONY3 should not produce a functional genome because it lacks the appropriate LTR sequences. Virus was produced by transient three plasmid cotransfection of 293T cells with pRV67, pONY3 and pONY2.10nlsLacZ as described for MLV-based vectors (Soneoka et al 1995 Nucl. Acids Res. 23:628-633) and then used to transduce 293T cells and D17 cells as follows. 48 hours post-transfection tissue culture fluid was collected and filtered through 0.45,um filters. Ten-fold dilutions were made in culture medium containing polybrene at 8 μg/ml and then 500 ul aliquots placed on D17 cells seeded at 1.6×105/well in 12 well plates on the previous day. Two hours later 1 ml of culture media was added and incubation continued for 48 hours prior to assessment of LacZ gene expression using the X-gal staining procedure. for E coli β-galactosidase (macGregor et al 1991 Methods in Molecular Biology Vol 7 ed EJ Murray p217-235). In both cases the virus transduced the cells at frequencies of about 105 LacZ-transducing cell—forming—unit (i.f.u.)/ml which was about 10-fold less than with the MLV-based vector produced from pH111. These data showed that we had produced an EIAV-based vector system and also suggested that replacement of the Hind III/Hind III fragment in env with foreign DNA may disrupt the function of the genome.
  • We next characterized the ability of the EIAV vector particles to be pseudotyped with envelope proteins from other viruses. pONY2.10nlsLacZ and pONY3 were cotranfected with envelope expression plasmids producing MLV amphotropic (pHIT456) and MLV ecotropic (pHIT123) envelopes (Soneoka et al 1995 Nucl. Acids Res. 23:628-633) as well as VSV-G (pRV67) (Table 1). pHIT111 (MLV vector genome) and pHIT60 (MLV gagpol expression plasmid) were cotransfected with the envelope plasmids as positive controls (Table 1). The viral supernatants were used to transduce a variety of cell lines including human kidney (293T), murine embryo (NIH3T3) and canine osteosarcoma (D17). As expected the cell tropism of the virus was largely determined by the envelope. EIAV could be pseudotyped with amphotropic envelope, but transduction efficiencies varied. The amphotropic pseudotyped virus gave titres of about 102 on D17 cells, 103 on NIH3T3 cells and 104 on 293T cells. The reason for these differences was not pursued. EIAV could also be pseudotyped with the MLV ecotropic envelope and these viruses transduced NIH3T3 cells at titres of 104 l.f.u./ml. EIAV, pseudotyped with VSV-G envelope, transduced all the cell lines tested. The titer varied between the different envelopes and cell types but overall efficiencies were relatively high for the non-murine cells, but still lower than with a murine vector system. Taken together, these data show that the EIAV vector system is not dependent on the EIAV envelope and can be effectively pseudotyped with three envelopes conferring broad host range. This makes this system as generally useful as current MLV-based systems. [0127]
  • EIAV vectors can also be pseudotyped in the same manner using the RD114 envelope, for instance using pRDF (Cosset et al 1995 J. Virol. 69: 7430-7436). [0128]
  • In order to characterize the transduction events further we carried out a PCR analysis of 293T cells transduced by the EIAV vector pseudotyped with VSV-G. In particular we asked if the vector genome, as opposed to a recombinant with the gagpol expression plasmid, pONY3, had been the transduction vehicle for the β-galactosidase gene. PCR amplification using primers specific for the EIAV LTR gave the expected PCR product of 310 bp when genomic DNA isolated from transduced cells was used (FIG. 2A, lane 1). No PCR product was detected when mock transduced 293T cell DNA was used as template (FIG. 2A, lane 2). pONY2.10nlsLacZ was used as a positive control (FIG. 2A, lane 3). No PCR product was detected when pONY3 was used as a template (FIG. 2A, lane 4). The lack of a PCR product, when using pol specific primers, (FIG. 2B) confirmed that no gagpol sequences from pONY3 had integrated into the host chromosomes. Taken together these data show that the authentic vector genome had transduced the cells. [0129]
  • In order to determine if the EIAV vector retained the ability to transduce non-dividing cells, 293T cells were arrested in G1/S phase by treatment with aphidicolin according to published procedures (Lewis and Emerman 1994) and then transduced with EIAV-based and MLV-based vectors pseudotyped with VSV-G. The transduction efficiency of the MLV vector was lower by four orders of magnitude in aphidicolin treated cells as compared to untreated cells. The incomplete block to cell transduction by MLV was probably due to a small population of dividing cells. In contrast, no significant difference was observed in the case of the EIAV-based vector. This demonstrates that the EIAV vector, like HIV vectors, can efficiently transduce non-dividing cells. The vector genome pONY2.10lacZ contains 1377 nt of gag. RNA secondary structure prediction (“http://www.ibc.wustl.edu/˜zuker/rna/”) was used to identify possible stem-loop structures within the leader and the 5′ end of gag. Based on these predictions four deletions were made within the gag region of pONY2.10lacZ (FIG. 1). Deletions were made by PCR mutagenesis using standard techniques. [0130]
  • pONY2.1lacZ contains 1377 nt of gag (deleted from position 1901 nt) [0131]
  • pONY2.11lacZ contains 354 nt of gag (deleted from position 878 nt) [0132]
  • pONY2.12lacZ contains 184 nt of gag (deleted from position 708 nt) [0133]
  • pONY2.13lacZ contains 109 nt of gag (deleted from position 633 nt) [0134]
  • pONY2.14lacZ contains 2 nt of gag (deleted from position 526 nt) [0135]
  • These vectors were used in a three plasmid cotransfection as described for MLV-based vectors (Soneoka et al 1995 Nucl. Acids Res. 23:628-633) and the virus generated was titred on 293T and D17 cells. [0136]
  • It was found that the first 109 nt of gag coding sequence were needed for maximal packaging in addition to the un-translated region; pONY2.13lacz (Table 2). Similar titres were found on D17 cells. The predicted secondary structure of the gag sequence derived RNA in pONY2.13lacZ is shown in FIG. 4. [0137]
  • Based on the secondary structure prediction in FIG. 4, four further deletions were made within the area upstream and downstream of the major splice donor codon in pONY2.13lacZ. [0138]
  • pONY2.2lacZ contains deleted between position 409 to 421 nt [0139]
  • pONY2.22lacZ contains deleted between position 424 to 463 nt [0140]
  • pONY2.23lacZ contains deleted between position 470 to 524 nt [0141]
  • pONY2.24lacZ contains deleted between position 529 to 582 nt [0142]
  • pONY2.25lacZ contains deleted between position 584 to 645 nt [0143]
  • pONY2.26lacZ contains deleted between position 409 to 421 nt and between position 470 to 542 nt. [0144]
  • These vectors were used in a three plasmid co-transfection as described above and the virus generated was titred on D17 cells. It was found that deletions within this region severely affected the titre of the virus (Table 3). Constructs pONY2.23 and 2.26 gave the lowest titre. These both contained the deletion between position 470 to 524 nt. The least severe deletion was the one between position 409 to 421 nt. Based on this information the region around the major splice donor is useful for optimal packaging. [0145]
  • Similar secondary structure predictions and deletion analysis may be used to identify the packaging signal in other non-primate lentiviruses. [0146]
    TABLE 1
    Transduction efficiency of viral vectors.
    Titer (l.f.u./ml)a
    Vector Envelope D17 NIH3T3 293T
    pONY2.1nlslacZ Mock <1 <1 <1
    pONY2.1nlslacZ pHIT456 1.0 × 102 8.4 × 102 2.0 × 104
    (MLVamp)
    pONY2.1nlslacZ pHIT123 <1 1.5 × 104 <1
    (MLVeco)
    pONY2.1nlslacZ pRV67 (VSVG) 1.0 × 105 3.6 × 103 2.0 × 105
    pHIT111 Mock <1 <1 <1
    pHIT111 pHIT456 1.3 × 105 2.6 × 106 2.0 × 107
    (MLVamp)
    pHIT111 pHIT123 <1 2.8 × 106 <1
    (MLVeco)
    pHIT111 pRV67 (VSVG) 3.0 × 106 2.0 × 105 5.0 × 106
  • [0147]
    TABLE 2
    Vector Titre
    Genome (l.f.u/ml)
    PONY2.10 3.30E + 04
    PONY2.11 1.60E + 05
    PONY2.12 1.40E + 05
    PONY2.13 1.70E + 05
    PONY2.14 5.40E + 02
    Mock  1.0E + 01
  • [0148]
    TABLE 3
    Vector Titre
    Genome (l.f.u/ml)
    2.21 1.20E + 04
    2.22 3.80E + 03
    2.23 1.20E + 02
    2.24 5.20E + 02
    2.25 5.60E + 02
    2.26 1.00E + 02
    2.13 4.00E + 04
  • EXAMPLE 2 Construction of pEGASUS-1
  • An EIAV—based vector was made (pEGASUS-1) that contains only 759 nt of EIAV sequences (268 nt-675 nt and 7942 nt-8292 nt) as follows. [0149]
  • Sequences encompassing the EIAV polypurine tract (PPT) and the 3′LTR were obtained by PCR amplification from pONY2.10LacZ using primers PPTEIAV+(Y8198): GACTACGACTAGTGTATGTTTAGAAAAACAAGG, and 3′NEGSpeI (Y8199):CTAGGCTACTAGTACTGTAGGATCTCGAACAG. The product was purified, digested with SpeI (ACTAGT) and ligated into pBS II KS+ which had been prepared by digestion with SpeI and treatment with alkaline phosphatase. Colonies obtained following transformation into [0150] E. coli, XL-1Blue were screened for the presence of the 3′LTR in the orientation in which the U5 region of the 3′LTR was proximal to the NotI site of the pBS II KS+linker. The sequence of the cloned insert was determined and showed that it contained only one change from the EIAV clone pSPEIAV19 (AC: U01866). This was a ‘C’ insertion between bases 3 and 4 of the R region. The same change was found in the template used in the PCR reaction. The clone was termed pBS.3′LTR.
  • Next the reporter gene cassette, CMV promoter/LacZ, was introduced into the PstI site of pBS.3′-LIR. The CMV/LacZ cassette was obtained as a PstI fragment from pONY2.10LacZ (see above). The ligation reaction to join the above fragments was transformed into [0151] E. coli, XL-1Blue. A number of clones in which the CMV/LacZ insert was orientated so that the LacZ gene was proximal to the 3′LTR were assessed for activity of the CMV/LacZ cassette by transfection into the cell line 293T using standard procedures. A clone which gave blue cells at 48 hours post-transfection following development with X-gal was selected for further use and termed pBS CMVLacZ.3′LTR.
  • The 5′region of the EIAV vector was constructed in the expression vector pCIEneo which is a derivative of pCIneo (Promega) modified by the inclusion of approximately 400 base pairs derived from the 5′end of the full CMV promoter as defined previously. This 400 base pair fragment was obtained by PCR amplifcation using primers VSAT1 (GGGCTATATGAGATCTTGAATAATAAAATGTGT) and VSAT2 (TATTAATAACTAGT) and pHFT60 as template. The product was digested with BglII and SpeI and ligated into pCIneo which had been digested similarly. [0152]
  • A fragment of the EIAV genome running from the R region to nt 150 of the gag coding region (nt 268—to 675) was amplified with primers CMV5′EIAV2 (Z0591)(GCTACGCAGAGCTCGTTTAGTGAACCGGGCACTCAGATTCTG: and 3′PSI.NEG (GCTGAGCTCTAGAGTCCTTTTCTTTTACAAAGTTGG) using as template DNA. The 5′region of the primer CMV5′EIAV2 contains the sequences immediately upstream of the CMV promoter transcriptional start site and can be cut with SacI. 3′PSI.NEG binds 3′ of the EIAV packaging sequences as defined by deletion analysis (above) and contains an XbaI site. The PCR product was trimmed with SacI and XbaI and ligated into pCIEneo which had been prepared for ligation by digestion with the same enzymes. This manipulation places the start of the EIAV R region at the transcriptional start point of the CMV promoter and transcripts produced thus start at the genuine start position used by EIAV and extend to the 3′-side of the packaging signal. Clones which appeared to be correct as assessed by restriction analysis were sequenced. A clone termed pCIEneo.5′EIAV was selected for further work. [0153]
  • In the next step the CMVLacZ and 3′LTR cassette in pBS.CMVLacZ.3′LTR was introduced into pCIEneo.5′EIAV. pBS.CMVLacZ.3′LTR was digested with ApaI, the 3′ overhangs removed with T4 DNA polymerase, then digested with NotI. The fragment containing the CMVLacZ.3′LTR was purified by standard molecular biology techniques. The vector for ligation with this fragment was prepared from pCIEneo.5′EIAV by digestion with SalI, followed by filling-in of the 5′ overhangs using T4 DNA polymerase. The DNA was then digested with NotI and purified prior to use in ligation reactions. Following transformation into [0154] E. coli, XL-1Blue colonies were screened for the presence of the insert by restriction analysis to identify the required clone, designated pEGASUS-1.
  • The function of the pEGASUS-1 EIAV vector was compared to pONY2.10LacZ using the three plasmid co-transfection system as described in Example 1. Comparable titres were obtained from both vectors indicating that pEGASUS-1 contains all the sequences required for packaging with good efficiency. [0155]
  • EXAMPLE 3 Introduction of RRE's into EIAV Vectors
  • Further improvements to the EIAV vector pEGASUS-1 may be made by introduction of additional elements to improve titre. A convenient site for the introduction of such elements is the SalI site which lies between the XbaI to the 3′ of the packaging signal and upstream of the CMV/LacZ cassette of pEGASUS-1. For example the RRE from HV or EIAV can be inserted at this site. [0156]
  • The HIV-1 RRE was obtained from the HIV-1 molecular clone pWI3 (Kimpton and Emerman 1992 (J. Virol. 66: 2232-2239) by PCR amplification using primers RRE(+) GTCGCTGAGGTCGACAAGGCAAAGAGAAGAG and RRE(−) GACCGGTACCGTCGACAAGGCACAGCAGTGG. The fragment of DNA and pEGASUS-1 were digested with SalI and following ligation, transformed into [0157] E. coli, XL-1 Blue. Colonies were screened for the presence of the HIV RRE and two clones, with the HIV RRE in either the positive or negative orientation, used for further work These vectors, pEGASUS-2.HIV RRE(+) or pEGASUS-2.HIV RRE(−) can be tested in 293T cells by carrying out a four plasmid co-transfection in which the plasmid pCIneoHIVrev, expressing the rev protein from HIV-1 is co-transfected with vector, pONY3 and pRV67 plasmids
  • The EIAV RRE as defined previously (Martarano et al 1994) was obtained by PCR amplification as follows. Using pONY2.10LacZ as [0158] template 2 amplifications were performed to obtain the two parts of the EIAV RRE. The 5′-element was obtained using primers ERRE1 (TTCTGTCGACGAATCCCAGGGGGAATCTCAAC) and ERRE2 (GTCACCTTCCAGAGGGCCCTGGCTAAGCATAACAG) and the 3′element with ERRE3 (CTGTTATGCTTAGCCAGGGCCCTCTGGAAGGTGAC) and ERRE4 (AATTGCTGACCCCCAAAATAGCCATAAG). These products will anneal to each other hence can be used in second PCR reaction to obtain a DNA which ‘encodes’ the EIAV RRE. The PCR amplification is set up with out primers ERRE1 and ERRE4 for the first 10 cycles and then these primers are added to the reaction and a further 10 cycles of amplification carried out. The resulting PCR product and pEGASUS-1 were digested with SalI, ligated and transformed into E. coli XL-1Blue. Clones in which the EIAV RRE was in either the positive or negative orientations were selected for further work. The activity of these vectors was assessed in 4-way co-transfectioand pEGASUS-1 were digested with SalI, ligated and transformed into E. coli XL-1Blue. Clones in which the EIAV RRE was in either the positive or negative orientations were selected for further work. The activity of these vectors was assessed in three palsmid co-transfections, (EIAV rev being supplied by pONY3) or in 4-plasmid co-transfection experiments as described above, but using pCIneo.EIAV Rev to supply additional EIAV rev.
  • For construction of pCIneo EIAV REV the EIAV REV encoding sequences were derived by PCR amplification. The EIAV REV sequences were obtained using a two step ‘overlapping’ PCR amplification procedure as described above for the EIAV RRE. Template for the two reactions was pONY3 and primers for the 5′fragment were [0159] EIAV REV REV 5′O(CCATGCACGTCTGCAGCCAGCATGGCAGAATCGAAG) and EAIV REV IN (CCTGAGGATCTATTTTCCACCAGTCATTTC) and for the 3′product EIAV REV IP (GTGGAAAATAGATCCTCAGGGCCCTCTGG) and EIAV.REV3′O (GCAGTGCCGGATCCTCATAAATGTTTCCTCCTTCG). The second PCR amplification was carried out with primers EIAV REV5′O and EIAV REV3′O being added after 10 cycles. The resulting product was ligated with the PCR fragment ‘TA’ cloning vector pCR2.1 (Invitrogen) the orientation of the EIAV REV insert was assessed by restriction enzyme analysis and the presence of the correct EIAV REV sequence confirmed. The construct was called pTopoRevpos. The EIAV REV insert was excised from pTopoRevpos by digestion with SpeI and NotI and ligated into pCIneo which had been digested with NheI and NotI.
  • EXAMPLE 4 Transduction of Human Macrophages
  • Primary human monocytes were obtained from leukocyte-enriched blood (from the National Blood Transfusion Service, Southmead Rd Bristol, UK) as follows. Peripheral blood mononuclear cells (PBMC) were enriched by centrifugation above a Ficoll discontinuous gradient (Pharmacia) according to the manufacturer's instructions Macrophages were obtained from this cell population by adherence to tissue culture plastic over 7 days in RPM! 1640 medium (Dutch modified; Sigma) containing 2% heat-inactivated human AB serum (Sigma) or 10% FCS (Sigma). Non-adherent cells were removed by extensive washing of the plates with medium. [0160]
  • Virus for transduction experiments was obtained by three plasmid co-transfection into 293T cells. The vector for the experiments was a pONY2.13 derivative in which the CMV/LacZ reporter cassette had been replaced with CMV/green fluorescent protein (GFP). [0161]
  • Vector pONY2.13GFP was made as follows. The sequence encoding the red-shifted GFP and eukaryotic translation signals was cut out of pEGFP-N1 (Clontech “http://www.clontech.com/”) with BglII and XbaI and ligated into the general cloning vector pSP72 (Promega) which had been prepared by digestion with the same enzymes. The GFP-encoding sequences were then excised using XhoI and ligated into pONY2.13 which had been cut with XhoI (thereby releasing the LacZ coding region). Following transformation into [0162] E. coli, XL-1Blue clones in which the orientation of the GFP insert with respect to the CMV promoter was such that expression would be expected were determined restriction analysis and expression of GFP confirmed by transfection of DNA into 293T cells.
  • Vector was recovered by three plasmid co-transfection into 293T cells and harvested at 42-48 hours post-transfection: tissue culture fluid was 0.45(m-filtered and virus was then pelleted by centrifugation at 50,000 g (20 Krpm), for 90 minutes at 4(C in a SW40Ti rotor. Virus was resupended in 50-100(1 of complete media for 2 hours and then used in transduction experiments. Transductions with pONY2.13GFP vector were carried out as follows. Macrophages, seeded at 5×10[0163] 5 per well of 48-well plates were washed once with medium and then 300 (l of medium was put back on the cells. Virus was added to the medium and gently pipetted up 2-3 times to ensure mixing. Transduction efficiency was assessed at 3-5 days post-transduction. The number of transduced macrophages was determined using a fluorescence microscope. Expression of GFP can be monitored for extended periods, e.g., up to several weeks. Alternatively, transductions can be carried out with vectors carrying the LacZ marker. In such experiments the transduction frequency is assessed by detecting the presence of β-galactosidase using immunological procedures.
  • EXAMPLE 5 Introduction of EIAV Vectors in vivo in Rat Brain
  • Adult Wistar rats were anaesthetised with a solution containing 1 part Nembutal (0.1 ml/35 gm body weight) 1 part Novetal (0.1 ml/35 gm body weight) and 2 parts dH2O, and placed into a stereotaxic apparatus. A midline incision was made along the rostral-to-caudal length of the scalp and the skin deflected back to expose the skull. Using stereotaxic coordinates (measured from Bregma) of 3.00 mm posterior and 3.00 lateral, a 1 mm diameter hole was drilled into the skull. Unilateral intracortical injections of EIAV vectors were then made using a 10 μl Hamilton syringe or a 1.0 μl fine glass capillary to various depths from the surface of the brain. The syringe was left in place an additional 5 min to prevent reflux. Control animals receive a single 10 μl intracortical injection of saline with the Hamilton or 1.0 μl with the fine glass capillary. Animals were then sutured and left to recover. Forty-eight hours later, these animals were deeply anesthetized as described above and perfused through the heart with 200 ml of phosphate-buffered saline (PBS). The brains were then dissected out, frozen into dry-ice cooled isopentane (−30° C.) and cut coronally at 10 μm with a cryostat. Every 5th section through the injection site and 2 mm rostral and caudal are collected onto Super-Frost slides, fixed and either X-gal or immunostained or stained with Cresyl Violet. [0164]
  • EXAMPLE 6 Transduction of Bronchial Cells Differentiated in Culture
  • Epithelial cells can be differentiated to form epithelia-like monolayers which display (>1000 Ω cm[0165] 3) electrical resistance and a cuboidal morphology. There are various wasys to do this for example Fuller et al 1984.This creates polarized cells. This polarity is functional and mimics epithelial cells in vivo. Thus EIAV vectors can be used to transduce these cells either through the basolateral surface or the apical surface using vectors and preparations as described in Examples 1-3.
  • EXAMPLE 7 A Minimal EIAV System
  • In order to eliminate the risk of accessory genes or coding sequences having deleterious effects in therapeutic applications, vector systems lacking tat, S2 and the dUTPase are constructed. [0166]
  • Construction of S2 Mutants [0167]
  • A) Vector Genome [0168]
  • pONY2.13lacZ contains 109 nt of gag (deleted from nucleotide positions 633 to 4949) (pONY2.13lacZ is described above). This vector is used to make an EIAV vector genome from which S2 expression is eliminated by deletion from nucleotide positions 5345 to 5397. This removes the ATG start codon of S2 and the start codon of env. To make the deletion within S2, PCR is carried out with SY2/SY5 and SY3/SY4 using pONY2.13 DNA as template. The two PCR products are pooled and PCR is carried out with primers SY5 and SY3. The 10.1 kb product is ligated into pGEMT-easy (Promega) to make pGEMS2 and sequenced to confirm the deletion. pONY2.13lacZDS2 is made by cutting out the 1.1 kb S2 region from pGEMS2 with Cel II and ligating it into pONY2.13lacZ. [0169]
  • B) Gagpol Construct [0170]
  • The same region of S2 is deleted in pONY3 to prevent recombination between pONY3DS2 and pONY2.13DS2 reconstituting the S2 gene. pONY3DS2 is made by PCR amplification with SY1/SY2 and SY3/SY4 using pONY3 DNA as template. The two PCR products are pooled and PCR is carried out with primers SY1 and SY3. The 0.7 kb product is ligated into pGEMT-easy (Promega) to make pGEMS22 and sequenced to confirm the deletion. The 0.7 kb S2 coding region is excised of pGEMS22 with Not I and inserted into pBluescript KS+(Stratagene) to make pBPCRS2. The Eco RV and Nco I fragment from pONY3 (2.2 kb) is inserted into pBPCRS2 cut with Eco RV and Nco I to make pBpONYS2. This is then cut with Eco RV and Cel II (2.9 kb fragment) and inserted into pONY3 cut with Eco RV and Cel II to thereby making pONY3DS2. [0171]
  • Construction of dUTPase Mutant [0172]
  • pONY3DdUTPase is made by site directed mutagenesis of nucleotide 4176 from a T to an A residue (Payne et al., Virology, 210:302-313). This mutates the aspartic acid to a glutamic acid. This is done by PCR amplification using PCR primers dUTPaseF and dUTPaseR. The template DNA is pONY[0173] 3. The PCR product is inserted into pGEMT-easy and sequenced to confirm the mutation. This is called pGDdUTPase. pONY3 is cut with Not I and Eco RV (4.6 kb) and inserted into pBluescript KS+(Stratagene) to make pBEV. The pGDdUTase is cut with Pac I and Pst I and the 0.4 kb band inserted into pBEV cut with Pac I and Pst I. This is called pONY3pBDUTPase. This is then inserted into pONY3 via N{overscore (o)}t I and Eco RV (4.6 kb) to make pONY3DdUTPase.
  • Construction of the S2 and dUTPase Double Mutant [0174]
  • To make the double mutant of pONY3 the construct pBpONYS2 is used. pGDdUTPase is cut with Pac I and Pst I and the fragment inserted into pBpONYS2 cut with Pac I and Pst I to make construct pS2DdUTPase. This is then cut with Eco RV and Cel II and inserted into pONY3 cut with Eco RV and Cel II to make pONY3DS2DdUTPase. [0175]
  • Analysis of S2 and dUTPase Mutants [0176]
  • pONY2.13lacZDS2, pONY3DdUTPase and pONY3DS2DdUTPase vectors are used in a number of combinations in three plasmid co-transfections to generate virus as described for MLV-based vectors (Soneoka et al 1995 Nucl. Acids Res. 23:628-633) and the virus generated is titred on 293T and D17 cells, in either dividing or non-dividing states. Cells are arrested in G[0177] 1/S phase by treatment with aphidicolin (9) and then transduced with EIAV-based and MLV-based vectors pseudotyped with VSV-G (Table 4). The transduction efficiency of the MLV vector is lower by four orders of magnitude in aphidicolin treated cells as compared to untreated cells. The incomplete block to cell transduction by MLV is probably due to a small population of dividing cells. In contrast, no significant difference is observed in the case of the EIAV-based vectors. This demonstrates that the EIAV-based system does not require S2 or dUTPase either for production or transduction. Payne et al., (Payne et al., Virology, 210:302-313) and others have shown that EIAV dUTPase is required for the infection of horse macrophages. This may represent a restriction in infection of macrophages by EIAV.
  • The properties of the S2 and dUTPase mutants are tested by transduction of hippocampal embryonic day 14 neuronal cells cultured in minimal media for 7 days. No significant difference is found between the various EIAV vectors. However a much reduced transduction efficiency is seen for the MLV vector. This indicates that S2 and dUTPase is not required for the transduction of physiologically non-dividing cells. [0178]
  • In summary we can conclude that tat, S2 and dUTPase are not required in any part of the vector system for vector production or transduction. [0179]
  • EXAMPLE 8 Addition of Rev/RRE
  • The construction of pEGASUS-1 has been described above. This vector contains 759 bp of EIAV sequence. The introduction of the EIAV RRE (0.7 kb) into pEGASUS-1 to produce pEGASUS/RRE resulted in a four-fold increase in the titre when Rev is provided in trans (Table 2). This vector now contains 1.47 kb of EIAV. [0180]
  • Example 9 Construction of Improved Gagpol Expression Plasmids
  • In pONY3 there is an extended 5′ untranslated region before the start of the gagpol coding sequence. It is likely that this unusually long sequence would compromise expression of the gagpol cassette. To improve gagpol expression pONY3 is modified to remove the remaining 5′ LTR. This is done by cutting pONY3 with Nar I and Eco RV. The 2.4 kb fragment is inserted into pBluescript KS+(Stratagene) at Cla I and Eco RV sites to make construct pBSpONY3.0. pBSpONY3.0 is cut with Xho I and Eco RV. The 2.4 kb fragment is inserted into pONY3 at Xho I and Eco RV to make pONY3.1. [0181]
  • This manipulation removes the 5′ LTR up to the Nar I site within the primer binding region (386 nt). This construct gives a two fold increase in titre and increased protein expression (FIG. 10). [0182]
  • pONY3.1 like pONY3 encodes gag, gagpol, Tat, S2 and Rev. Since the S2 mutation experiments showed that S2 is not required either in the production system or in the EIAV vector genome it is possible to design a gagpol expression constructs without S2. Two such constructs, pHORSE and pHORSE3.1, are produced. [0183]
  • pHORSE is made by PCR amplification with EGAGP5′OUTERIEGAGPINNER3 and EGAGP3′OUTER/EGAGPINNER5 using pONY3 as template DNA. The two PCR products are purified pooled and re-amplified using primers EGAGP5′OUTER/EGAGP3′OUTER. This product is inserted into the Xho I and Sal I sites of pSP72 to make pSP72EIAVgagpolO'lap. [0184] pONY 3 is cut with Pvu II and Nco I and the 4.3 kb fragment is inserted into pSP72EIAVgagpolO'lap cut with Pvu II and Nco I to make pSPEGP. This is cut with Xho I and Sal I (4.7 kb) and inserted into pCI-Neo at the Xho I and Sal I sites. This construct is called pCIEGP. The RRE is cut out from pEGASUS with Sal I (0.7 kb) and inserted into pCIEGP construct at the Sal I site to make pHORSE.
  • When this construct is assayed for protein expression in the presence or absence of pCI-Rev (a construct expressing the EIAV Rev open reading frame, see above) it is found to be Rev dependent as expected. However, protein expression is much lower than from pONY3.1. In addition when used in virus production the titre is found to be 100 fold lower than that from pONY3.1. [0185]
  • Unexpectedly when the leader sequence (comprising sequences from the end of U5 of the 5′ LTR to the ATG start of gag 383-524 nt) of pONY3.1 is inserted into pHORSE, to make pHORSE3.1, protein expression and virus production improved. pHORSE3.1 is made by replacing the 1.5 kb Xho IXba I of pHORSE with the 1.6 kb Xho I/Xba I of pONY3.1. Titres obtained with pHORSE3.1 are similar to that of pONY3.1. The reason for the slightly lower titre of pHORSE3.1 compared to pONY3.1 may be due the requirement for a four plasmid co-transfection with pHORSE3.1 (due to the Rev dependence of this system). We can conclude therefore that a minimal EIAV vector system should have this leader for maximum gagpol expression. [0186]
  • When pHORSE3.1, pRV67, pCIRev and pEGASUS/RRE are used in a four plasmid co-transfection (Table 6) virus is produced at a high titre (2.0×10[0187] 4 l.f u./ml). This system lacks the second exon of Tat which is responsible for Tat transactivation (Southgate et al., J. Virology, 1995, 69:2605-2610). This demonstrates that the Tat is not required for the EIAV-based vector system.
  • By engineering the backbone of pHORSE3.1 to express Rev (replacing the Neo open reading frame with that of EIAV Rev) the requirement of a four plasmid co-transfection was eliminated. This was done by cutting pCI-Neo with Stu I and Bst XI and filling in the 5′ overhangs with T4 DNA polymerase. This produced a vector fragment of 4.6 kb into which the Rev open reading frame from pTopoRevpos (cut with Sac I and Xba I giving a 0.6 kb band in which the 5′ overhangs were filled in using T4 DNA polymerase) was inserted. This was called pCREV. The EIAV gagpol reading frame including the RRE and leader was cut from pHORSE3.1 with Xho I and Not I (5.5 kb) and inserted into pCREV at the Xho I and Not I sites to make pEGPR3.1. [0188]
  • Codon optimisation of the EIAV gagpol should eliminate the dependence of gagpol protein expression on the RRE/Rev system. The need of pEGASUS-1 for Rev/RRE can also be eliminated by using a heterologous RNA export system such as the constitutive transport element (CTE) from Mason-Pfizer Monkey virus (MPMV) (Bray et al., PNAS, 1994, 91:1256-1260, Kim et al., 1998) [0189]
    TABLE 4
    Titre on Ratio
    D17 cells (Non-
    (l.f.u./ml) Non- dividing/
    Vector gagpol Dividing dividing dividing)
    S2+ S2+, dUTPase+ 2.2 × 105 1.1 × 105 0.5
    S2− S2+, dUTPase+ 1.5 × 105 1.3 × 105 0.9
    S2− S2−, dUTPase+ 1.0 × 105 1.2 × 105 1.2
    S2− S2−, dUTPase− 1.5 × 105 1.6 × 105 1.1
    S2+ S2−, dUTPase+ 2.2 × 105 2.3 × 105 1.0
    S2+ S2−, dUTPase− 1.5 × 105 1.4 × 105 1.0
    S2+ S2+, dUTPase− 1.5 × 105 1.4 × 105 1.0
    MLV Vector 1.2 × 107 6.7 × 103 0.0006
    Mock <1 <1 1
  • [0190]
    TABLE 5
    Comparison of pONY2.10LacZ and pEGASUS +/− EIAV RRE.
    Titre
    Vector Genome Gagpol (l.f.u./ml)
    pONY2.10LacZ pONY3.0   7 × 104
    pEGASUS pONY3.0 2.2 × 104
    pEGASUS/RRE pONY3.0 8.6 × 104
  • Titres with Rev are higher for pEGASUS-1 even though it has no RRE. Possibly the effect of REV is via enhanced expression of gagpol. [0191]
    TABLE 6
    Titre
    Vector Genome Gagpol (l.f.u./ml)
    pONY2.11lacZ pONY3.1 1.7 × 105
    pONY2.11lacZ pHORSE3.1 9.0 × 104
    pEGASUS/RRE pONY3.1 8.0 × 104
    pEGASUS/RRE pHORSE3.1 2.0 × 104
  • Transfections were carried out in 293T cells with pCI-Rev and pRV67. The virus was titred on D17 cells [0192]
    Primers
    Figure US20030147907A1-20030807-C00001
    Figure US20030147907A1-20030807-C00002
    Figure US20030147907A1-20030807-C00003
  • EXAMPLE 10 pONY4 Series of Vectors
  • In order to eliminate the use of Tat for the transcription of the EIAV genome and increase the amount of full length transcript the EIAV U3 (5′ LTR) was replaced with the HCMV enhancer/promoter as in the case of the pEGASUS vectors (Example 2). [0193]
  • Plasmid Construction. [0194]
  • pONY2.1lacZ contains a deletion in gag such that only 373 bp of the gag ORF remains. pONY4 was made by replacing the 5′ LTR with the CMV LTR from pEGASUS-1. pEGASUS-1 was cut with Bgl II/Xho I releasing a 3.2 kb fragment (containing the CMV LTR) which was inserted into pSP72 cut with Bgl I/Xho L This construct was named pSPPEG213. This was cut with Hpa I/Nar I and the 1.3 kb fragment (encompassing the CMV LTR) was inserted into pONY2.11lacZ cut with Nae I/Nar L pONY4.1 contains a deletion (20.1 kb) downstream of the lacZ gene (between the Sfu I and Sal I sites) such that tat, S2, env, rev and RRE, are either missing or severely truncated (FIG. 11[0195] c). pONY4.1 was made by cutting it with Sfi I/Sal I, blunt-ended by Klenow polymerase and religated. pONY4G was made by replacing the lacZ gene of pONY4 (Sac II/Kpn I and then blunting with Klenow polymerase) with that of GFP from pEGFP-N1 (Clontech) (Bam HI/Xba I and then blunting with Klenow polymerase) as a blunt fragment.
  • Production and Assay of Vectors. [0196]
  • Vector stocks were generated by calcium-phosphate transfection of human kidney 293T cells plated on 10 cm dishes with 16 μg of vector plasmid, 16 μg of gag-pol plasmid and 8 ug of envelope plasmid. 36-48 h after transfection, supernatants were filtered (0.45,um) aliquoted and stored at −70° C. Concentrated vector preparations were made by ultracentrifugation of at 20 000 rpm (SW40Ti rotor) for 90 min, at 4° C. The virus was resuspended in PBS for 3-4 h aliquoted and stored at −70° C. Transduction was carried out in the presence of polybrene (8 μg/ml). It was consistently observed that pONY2.11 lacZ gave about 2 to 4 fold higher titres than the less deleted pONY2.10lacZ. When U3 in the 5′ LTR was replaced with the CMV enhancer/promoter as in pONY4 then titres increase a further 5 to 10 fold. [0197]
  • EXAMPLE 11 EIAV ‘Self-Inactivating’ Vectors (SIN-Vectors)
  • The expression of the transgene from EIAV vectors in particular cell types may be influenced by elements in the LTR's. To remove such elements SIN (Self Inactivating) vectors can be constructed however the precise configuration of the vector may be influenced by the requirement to maintain certain sequences necessary for efficient production of the vector (Mol Cell Biol 1996 Sep; 16(9):4942-51. RNA structure is a determinant of poly(A) site recognition by cleavage and polyadenylation specificity factor. Graveley B R, Fleming E S, Gilmartin G M) (J Virol 1996 Mar; 70(3):1612-7. A common mechanism for the enhancement of [0198] mRNA 3′ processing by U3 sequences in two distantly related lentiviruses. Graveley B R, Gilmartin G M). In addition SIN vectors provide a way for eliminating the production of full length transcripts in transduced cells.
  • Two SIN vectors were made: one containing the putatively important sequences (for polyadenylation), located between the Mlu I and Mun I sites and one in which these sequences were deleted. The 5′ border of the deletions was 112 bases from the 5′ end of the U3 region of the 3′LTR. The structure of two SIN vectors is shown in FIG. 12. [0199]
  • Deletions present in pONY4G.SIN-MLU and pONY4G.SIN-MUN vectors are indicated in dashed lines. Primers are shown in italic. [0200]
  • DNA sequences between nucleotides 7300 and 8079 (numbered according to EIAV clone pSPEAIV19, Accession No. U01866) were obtained using polymerase chain reaction amplification using pONY4G as template. The positive sense primer was ERRE3 and the negative primers for amplification were SIN-MLU (C7143: GTCGAGCACGCGTTTGCCTAGCAACATGAGCTAG (MluI site in bold) or SIN-MUN (C7142: GTCGAGCCAATTGTTGCCTAGC AACATGAGCTAG (MunI site in bold) where the underlined sequences are complimentary to nucleotides 8058 to 8079 (of pSPEIAV19). The PCR products were digested with NspV and either MuI or MunI respectively. These were then ligated into pONY4G prepared for ligation by digestion with NspV(SfuI) and either MluI (partial digestion) or MunI respectively. [0201]
  • EXAMPLE 12 EIAV Vectors with Reverse Configuration Internal Promoter-Reporter Cassettes
  • In EIAV vectors such as pONY4Z or pONY4G the internal CMV-reporter cassette is orientated so that transcription from the 5′LTR and the internal promoter are co-directional and the polyadenylation signal in the 3′LTR is used for transcripts from both promoters. An alternative configuration is achieved by reversing the internal promoter-reporter cassette, however a polyadenylation signal must be placed downstream of the cassette. [0202]
  • An example of this ‘reverse orientation’ vector was made as follows. pONY4Z was digested with PstI and the overhanging termini trimmed back with T4 DNA polymerase. This was then used as the ‘vector’ fragment in a ligation with the MluI to AseI fragment from pEGFP-C1 which contains sequences including the CMV-GFP-SV40 early mRNA polyA signal cassette. Prior to ligation this fragment was flush-ended with T4 DNA polymerase. The vector encoding plasmid was called pONY4Greverse. [0203]
  • Vector particles were recovered from pONY4Greverse by co-transfection with pONY3.1 and pRV67, which express EIAV gag/pol and VSV-G protein respectively. The titre on D17 canine cells from pONY4Greverse was 13-fold lower than from pONY4G vector recovered in parallel. [0204]
  • The lower titre of pONY4Greverse was probably due to interference between the CMV promoters which drive transcription of the genome and the GFP towards each other however truncation of the genomic RNA by the SV40-derived polyadenylation signal present in the inserted CMV-GFP-polyA cassette could also have been a factor. An improved vector was made by replacing the polyadenylation signal of pONY4Greverse with the bovine growth hormone polyadenylation (BGHpA) signal. To make this improvement pONY4Greverse was digested with BstAPI and the ends flushed with T4 DNA polymerase, then cut with PstI. This ‘vector’ fragment was then ligated to a DNA fragment representing the BGHpA which was prepared from pcDNA3.1+ (Invitrogen) by digestion with SphI, and then the ends blunted with T4 DNA polymerase, then digested with PstI. [0205]
  • EXAMPLE 13 Construction and use of poly.A Signals Containing Introns
  • In the pONY vectors described here the polyadenylation signal used is that from EIAV. This is found in the 3′ LTR at the border of R and U5. This signal may not be optimal because it is not of a consensus sequence (see Whitelaw and Proudfoot 1986 [0206] EMBO 5; 2915-2922 and Levitt et al 1989 Gen. and Dev. 3; 1019-1025 for description of consensus polyadenylation signal).
  • One method of improving the viral polyadenylation is to replace the 3′ LTR poly A signal with that of a consensus/strong polyadenylation signal. By such a method the signal would now be optimal in the producer cell. However upon transduction this signal is lost because during replication, the 5′ LTR is the source of the poly A signal (see Retroviruses 1998 CSH press (Ed. J. Coffin) for review of retroviral life cycle). One novel way of overcoming the problem (of no strong polyadenylation signal upon transduction) is to include the poly A signal in a manner as will now be outlined: The method is to use a ‘split poly-A signal’ where by an intron splits the aataaa motif from that of the essential g/u box. Such a signal has previously been used by Liu et al (1993 N.A.R 21;5256-5263) to demonstrate both that large gaps between the aataaa and the g/u box will disable the poly A signal and that the polyadenylation process preceeds splicing. By placing a split-polyA signal within the retroviral vector such a signal will not be functional until transduction of target cells. This is because polyadenylation preceeds splicing and as such the upstream split-polyA signal will not be used during vector expression within the producer cell. Outlined in FIG. 13 is a schematic representation of how such a retroviral vector, containing a split polyA signal, would function—both in producer and in transduced cells. First this Figure demonstrates that although there exists an upstream consensus polyadenylation signal, the initial vector transcripts are still polyadenylated at the usual 3′ LTR using either a viral or other poly A signal as so desired. This is because although the upstream poly A signal is functional in the final vector genome, this signal is not read by the polyadenylation machinery because it is created only during intron removal and thus not present in the primary RNA transcript. Second, this figure demonstrates that upon transduction the-resulting vector transcripts are now polyadenylated at the first signal; this being now a normal strong polyadenylation signal with no introns to distance the essential aataaa and g/u box. [0207]
  • There are a number of advantages to inclusion of such a split-poly A signal within a retroviral vector; these include the following: [0208]
  • (1) The use of strong non-viral based polyadenylation signal within the transduced cell will enhance gene expression within such cells. [0209]
  • (2) The use of such poly A signals upstream of the natural LTR (see FIG. 13) based signals will, upon transduction, generate shorter RNA transcripts that contain less viral sequence at their 3′ end and as such will not be able to undergo subsequent retroviral reverse transcription. Indeed if the desired gene is expressed from an internal promoter such as the CMV, rather than an LTR; the resulting transcript expressed in the transduced cell could be designed to contain no viral sequence at all (see FIG. 3A). [0210]
  • (3) Inclusion of such a signal upstream of the 3′LTR will mean expression of the RNA downstream to the split poly A signal will be limited only to the producer cell because such RNA will not be transcribed in the transduced cell. This will therefore restrict certain sequence expression (for example IRESneo; see FIG. 14B) to producer cells. [0211]
  • (4) The presence of an intron within the producer cell will help with nuclear export of vector RNA from the nucleus. [0212]
  • (5) Because upon transduction their now exists an internal functional poly A signal, the viral poly A signal in the 5′ LTR (the one copied to the 3′ position during reverse transcription) can be removed/deleted if desired. This is of use for preventing the process of promoter-proximal polydenylation from the 5′ LTR in the producer cell (see Scott and Imperiale 1997 (Mol. Cell. Biol. 17;2127-35) and thus encourage full length transcript production of the virus. [0213]
  • EXAMPLE
  • To demonstrate the use of such a signal in a retrovirus; the “split poly A signal” cassette is constructed as described in FIG. 15; with the intronic sequence being derived from pCI (Promega). Once made this cassette is cloned into the [0214] pONY 4 GFP vector using the PstI compatible unique sse8387 site of pONY4-GFP (see FIG. 16). Upon transduction the resulting vector will now polyadenylate prior to the 3′LTR and consequently no viral RNA 3′ to lacZ will be transcribed (see FIG. 16).
  • EXAMPLE 14 Construction of MLV/EIAV Vectors
  • By replacing the EIAV LTR sequences with the MLV equivalents, the pONY vectors will no longer possess functional tar elements within the repeat regions (R) and as a consequence the U3 promoter will function without the requirement of Tat in the transduced cell. [0215]
  • Outlined in FIG. 17 is how such a vector is made by overlapping PCR with primers described in FIG. 18. Primers Mel and Me2 are used to amplify a PCR product from the MLV vector pHIT111 (Soneoka et al 1995 [0216] NAR 23;628-633) whilst Me3 and Me4 are used to amplify a product from pONY4 lacZ. The resulting two products are then combined in a primerless PCR reaction to overlap them (homology between the two products is shaded in FIG. 17). The final full length product is cut BglII and Xbal and used to replace the BglII-Xbal fragment of pONY4 lacZ (containing the CMV/R/U5) to make pONY4-5′MLV. The resulting vector now has the CMV/R/U5 sequence from MLV linked to the EIAV U5 sequence (sequence required for genome recognition by intergrase prior to intergration). The next step involves PCR amplification with primers Me5 and Me6 from pONY4 LacZ template and PCR amplification with Me7 and Me8 from pLXSN template (Miller and Rosman 1989 Biotechniques 7:980-990). These two PCR products are then overlapped by primerless PCR (homology between primers shown as hatched box) and the resulting fragment cut with NspV and MunI and inserted into the NspV/MunI sites of pONY4-5′MLV; thus replacing the 3′EIAV LTR with a 3′MLV LTR fused to the 3′UTR/ppt/U3 intergrase binding site of pONY 4 lacZ. The resulting plasmid, named pONY-MOUSE (see FIG. 19 for complete DNA sequence), titres at 104-5 per ml when combined with pONY3.1 and pRV67 in the HIT system.
  • EXAMPLE 15 Early Promoter Driving Lentiviral Vector Genome
  • In this example an EIAV genome is expressed from a vaccinia early promoter P7.5E (Davison 1989a). The promoter has been engineered to produce an EIAV genome with the correct 5′ RNA end. In addition the vaccinia early termination sequence has been inserted downstream of the EIAV genome. This is inserted into the transfer plasmid pSC65, which can homologously recombine into the TK region of the MVA genome. Recombinant viruses can be selected by their lack of sensitivity to BudR (Earl et al. 1998). [0217]
  • FIG. 11 is a schematic representation of the EIAV genome vectors pONY4.0 and pONY4.1 which have been described in Example 10 and the vaccinia transfer vector pSC65 (Chakrabarti et al 1997). The P7.5E sequence is AAAAGTAGAAAATATATTCTAATTTATT. The Early termination sequence for the early promoters is TTTTTNT (N=any nucleotide) (Fields). [0218]
  • The DNA manipulations are as follows and FIGS. 20 and 21 give the sequence of the PCR primers. PCR with primers EMVA1/2 produces the 5′ LTR with the U3 region replaced by the P7.5E promoter. This is inserted into the plasmid pSP72 (Promega) using the Hind III/Pst I sites to make pEMVA1. EIAV U3 contains a sequence matching the criteria for vaccinia early termination (TTTTTAT). Using primers EMVA3/4 and EMVA5/6 and overlapping PCR this region is mutated to TTTCCAT in order to prevent early termination. This PCR product is inserted into the pEMVA1 using the Bgl II/Pst I sites to generate pEMVA2. A termination sequence (TTTTTTTTT) is inserted downstream of the 3′ LTR R region using primers EMVA7/8 . This PCR product is inserted into pEMVA2 using the Mun I/Bgl II sites making pEMVA3. Into this plasmid the rest of the EIAV vector genome (pONY4) is inserted via the Nar I/Nsp V sites making pEMVA4 (FIG. 22). This is then cut with Pac I/Bgl II and inserted into pSC65 cut with Pac I/Bam HI to make pEPONY4 (Bgl II and Bam HI are compatible) (FIG. 23). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65. [0219]
  • In order to make the minimal EIAV genome version of this construct that is analogous to pONY4.1, pEMVA4 is cut with Sal I/Nsp V blunt ended and religated to make pEMVA5 (FIG. 24). This removes much of the sequence between the end of the lac Z gene and the end envelope region, hence this vector is Tat, Rev, S2 and Env minus. This is described in Example 10. This is then cut with Pac I/Bgl II and inserted into pSC65 cut with Pac I/Bam HI to make pEPONY4.1 (Bgl II and Bam HI are compatible) (FIG. 24). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65. [0220]
  • Both pEPONY4.0 and pEPONY4.1 are suitable for inserting the genome expression cassettes into the TK region of the MVA genome (Carroll MW and Moss B Virology Nov. 24, 1997;238(2):198-211) using a BHK TK-ve cell line (ECACC 85011423) and standard procedures for the construction of recombinant poxviruses (Earl et al 1998a & 1998b) [0221]
  • EXAMPLE 16 Synthetic Early/Late Promoter Driving Lentiviral Vector Genome
  • The synthetic early/late promoter of vaccinia has a requirement for sequences downstream of the RNA initiation site (Davison 1989b). For this promoter to be used to generate a retroviral genome either the R regions have to be modified or a ribozyme is used to make the correct 5′ end. Modifying the R regions is problematic as the initiation site has not been conclusively identified and varies with the sequence (Davison 1989b). Below is described the generation of a transfer plasmid that expresses the EIAV genome from the synthetic early/late promoter (Psyn). Downstream of this promoter is inserted a ribozyme that ensures the creation of the correct 5′ end of the RNA. This construct also contains the early termination sequence. [0222]
  • The DNA manipulations are as follows: PCR with primers EMVA9/1 produces the 5′ LTR with the U3 region replaced by the Psyn promoter and a hammerhead ribozyme (FIGS. 25 and 26). This is inserted into the plasmid pEMVA4 (Example 15) using the Pac I/Nar I sites to make pEMVA6 (FIG. 27). This is then cut with Pac I/Bgl II and inserted into pSC65 cut with Pac I/Bam HI to make pSynPONY4 (Bgl II and Bam HI are compatible) (FIG. 27). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65. In order to make the minimal ELAV genome version of this construct that is analogous to pONY4.1, pEMVA6 is cut with Sal I/Nsp V blunt ended and religated to make pEMVA7 (FIG. 28). This is then cut with Pac I/Bgl 11 and inserted into pSC65 (a vaccinia transfer vector) cut with Pac I/Bam HI to make pSynPONY4.1 (Bgl II and Bam HI are compatible) (FIG. 28). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65. [0223]
  • Both pSynPONY4.0 and pSynPONY4.1 are suitable for inserting the genome expression cassettes into the TK region of the MVA genome (Carroll MW and Moss B Virology Nov. 24, 1997;238(2):198-21 1) using standard procedures for the construction of recombinant poxviruses (Earl et al 1998a & 1998b) [0224]
  • EXAMPLE 17 T7 Promoter Driving Lentiviral Vector Genome
  • The T7 promoter can be used to generate a retroviral genome which can make the correct 5′ end. Below is described the generation of a transfer plasmid that expresses the EIAV genome from the T7 promoter (T7). Downstream of this promoter is inserted a T7 termination sequence. This is inserted into the transfer plasmid pSC65, which can homologously recombine into the TK region of the MVA genome. The T7 promoter requires the T7 polymerase. MVA viruses are available which express T7 polymerase from Vaccinia promoters (Wyatt et al 1995). [0225]
  • The T7 promoter has the sequence (−)TAATACGACTCACTATAGG(+2) with transcription beginning after A with preferably a run of Gs. The T7 termination sequence is CTAGCATAACCCCTTGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTG. The T7 promoter and terminator sequences are as those described in the plasmid pCITE-4a(+) (Novagen). [0226]
  • The DNA manipulations are as follows. PCR with primers EMVA10/11 (FIG. 29) produces the 5′ LTR with the U3 region replaced by the T7 promoter. This is inserted into the plasmid pEMVA4 using the Pac liNar I sites to make pEMVA8 (FIG. 30). PCR with [0227] primers EMVA 1/7 produces part of the 3′ LTR with a T7 termination sequence. This is inserted into pEMVA8 using the Mun I/Bgl II sites to make pEMVA9. This is then cut with Pac I/Bgl II and inserted into pSC65 (a vaccinia transfer vector) cut with Pac I/Bam HI to make pT7PONY4 (Bgl II and Bam HI are compatible) (FIG. 30). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65.
  • To make the minimal EIAV genome version of this construct (pONY4.1) pEMVA9 is cut with Sal I/Nsp V blunt ended and religated to make pEMVA10 (FIG. 31). This is then cut with Pac I/Bgl II and inserted into pSC65 (a vaccinia transfer vector) cut with Pac I/Bam HI to make pT7PONY4.1 (Bgl II and Bam HI are compatible) (FIG. 31). This removes the two vaccinia promoters and the lacZ coding cassette from pSC65. [0228]
  • Both pT7PONY4.0 and pT7PONY4.1 are suitable for inserting the genome expression cassettes into into the TK region of the MVA genome (Carroll MW and Moss B Virology 1997) using standard procedures for the construction of recombinant poxviruses (Earl et al 1998a & 1998b). [0229]
  • EXAMPLE 18 Construction of an EIAV Gagpol Cassette for Expression in Vaccinia
  • Normally EIAV gag/pol requires Rev/RRE for expression as Rev enables the unspliced transcript to be exported out of the nucleus. As Pox viruses are cytoplasmic, EIAV viral RNA export should not be a problem. But if Rev has other functions such as RNA stability or acts as a translation enhancer it can be expressed in a similar way to EIAV gag/pol (Martarano 1994). Alternatively the EIAV gag/pol sequence can be codon optimised to overcome the Rev/RRE requirement for export and enhance RNA stability. Below is described the creation of a vector that expresses EIAV gag/pol from a synthetic early/late promoter (Psyn). This is inserted into the transfer plasmid pLW-22 (Wyatt and [0230] Moss Appendix 1, Earl et al 1998a & b), which can homologously recombine into the Del II region of the MVA genome. Recombinant viruses can be selected by their expression of lac Z.
  • EIAV gag/pol including the leader the gag/pol open reading frame and the RRE can be obtained from cutting pHORSE3.1 (Example 9) with Aho LINot I to give a 5.5 kb band (FIG. 32). This is then inserted into the vaccinia transfer vector pLW-22 cut with Sal I/Not I (Sal I and Xho I are compatible) to make pLWHORSE3.1 (FIG. 32). [0231]
  • EXAMPLE 19 Construction of an EIAV Rev Cassette for Expression in Vaccinia
  • In the event that Rev is required for EIAV viral vector production from a poxvirus it can be expressed from a synthetic early/late promoter. This construct is inserted into the transfer plasmid pMC03, which can homologously recombine into the Del III region of the MVA genome. Recombinant viruses can be selected by their expression of GUS (Carroll et al. 1995). [0232]
  • The DNA manipulations are as follows. Plasmid pCIRev is described in Example 9. It is an EIAV Rev expression plasmid. This is cut with Afl II/Not I (0.6 kb), blunt ended by T4 DNA polymerase and inserted into pMC03 (Carroll et al. 1995) cut with Pme I to make pMCRev (FIG. 33). [0233]
  • EXAMPLE 20 Construction of Heterologous Envelope Cassettes for Expression in Vaccinia
  • EIAV can be pseudotyped with a number of envelopes such as VSV-G and amphotropic MLV envelope. Below is described the creation of a MVA transfer vector that expresses the amphotropic envelope or VSV-G envelope from the P7.5 early/late promoter. The transfer vector is pYF6 which can homologously recombine into the HA region of MVA. Recombinant viruses can be selected by direct live immunostaining for expression of the env. [0234]
  • In order to produce a transfer vector containing a VSV-G cassette, the VSV-G expression plasmid pRV67 (Kim et al. 1998) is cut with Sma I/Eco RV(1.7 kb) and the resulting fragment inserted into pYF6 cut with Sma I to make pYFVSVG (FIG. 34). Similarly, to produce an analogous amphotropic envelope construct pHIT456 (Soneoka 1995) is cut with Xba I and the 2.2 kb band blunt ended by T4 DNA polymerase and inserted into pYF6 cut with Sma Imaking pYFAmpho (FIG. 35). [0235]
  • Both pYFAmpho and pYFVSVG are suitable for inserting the genome expression cassettes into into the HA region of the MVA genome using standard procedures for the construction of recombinant poxviruses (Earl et al 1998a & b, Flexner et al 1987) [0236]
  • EXAMPLE 21 Construction and Amplification of MVA-Lenti Recombinants
  • The recombinant vaccinia viruses containing multiple inserts encoding the components of the EIAV vectors (FIG. 25) are constructed by sequential recombination with the relevant transfer plasmids. The construction of v.MEeG-Or (FIG. 36) is used as an example: [0237]
  • 1. A plasmid carrying gag-pol (pLWHORSE3.1) is transfected into BHK-21 or CEF cells, that have been previously infected with MVA (as described in Carroll and Moss 1997, Earl et al 1998a & b). [0238]
  • 2. After two days of infection recombinant MVA virus is assayed on BHK-21/CEF and cells are over-layed with agar medium containing the substrate for the colour marker P-galactosidase (Chakrabarti et al 1985) which is expressed from within pLW22. [0239]
  • 3. Blue plaques are picked and plaque purified until a homogeneous recombinant virus population is obtained. [0240]
  • 4. Recombinant virus is then used to recombine with transfer plasmids containing the other recombinant genes: pMCRev in which selection is based on GUS expression (Carroll & Moss 1995), the genome (pEPONY4.0) in which selection is based on a TK negative phenotype using BudR (Carroll & Moss 1997, Earl et al 1998a & b) and VSVG (PYFVSVG) in which recombinants are identified by direct immunostaining of VSV G (Earl et al 1998a & b). [0241]
  • Recombinant viruses may be amplified in BHK-21 or CEF cells as described below: [0242]
  • Propagation of Vaccinia Virus [0243]
  • The highly attenuated strain MVA is derived from the replication competent strain Ankara and has endured over 570 passages in primary chick embryo fibroblast cells. MVA replication was initially thought to be restricted to CEF cells as only minimal replication in mammalian cells was reported. However, further analysis has shown that Baby Hamster Kidney cells (BHK-21) are able to support high titre production of MVA. MVA may thus be grown on BHK-21 or primary CEF cells (Carroll & Moss (1997) Virology 238:198-211). [0244]
  • To prepare CEF cells, 10 day old chick embryos are gutted and limbs and head are removed before being minced and trypsinised in a solution of 0.25% trypsin and incubation at 37° C. The cell suspension is filtered through a course filter mesh and cells are washed and concentrated by centrifugation at 2000 rpm in a Sorvall RC-3B at 1500 rpm for 5 mins. Cells are suspended in MEM containing 10% FCS, aliquotted into 175 cm flasks and incubated at 37° C. in a CO[0245] 2 incubator. When monolayers are 95% confluent they are trypsinised and used to seed additional flasks or six well plates. Alternatively, primary cultures are transferred to a 31° C. incubator for later use (Sutter and Moss (1992) Proc Natl Acad Sci U S A 89:10847-10851).
  • Preparation of Crude, Semi-Purified and Purified Virus Stocks [0246]
  • Crude virus stocks are prepared for initial recombinant virus analysis or as viral stocks used for subsequent high titre virus preparations. Vaccinia virus preparations can be semi-purified by centrifuging out cell membranes and nuclei or by additional steps involving sucrose centrifugation to prevent contamination by pre-expressed recombinant protein products and cellular organelles. Methods used are a modification of those described by Earl et al., 1998a & b; Earl and Moss, ibid, pp. 16.17.1-16.17.16; Earl and Moss, ibid, pp. 16.18.1-16.18.10; and Bronte et al., (1997) Proc Natl Acad Sci U S A 94(7):3183-3188. [0247]
  • Crude Virus [0248]
  • MVA is grown in either CEF or BHK-21 (obtained from the ATCC) and WR is grown in HeLa or BS-C-1 (ATCC) in 175 cm[0249] 2 tissue culture flasks. Briefly, confluent monolayers are infected with an moi of approx. 1 pfu with MVA or WR. Virus is suspended in 10 ml MEM containing 2% FCS and added to 175 cm2 flasks containing confluent cell monolayers. After inoculation for 1 hour at 37° C. an additional 20 ml MEM containing 2% FCS is added. After 48-72 hours infected cells are scraped into the medium and pelleted at 1500 g for 5 mins. For crude virus preparations cells are resuspended 2 ml MEM (2%) per 175 cm2 flask. Cells are freeze thawed three times, sonicated and aliquotted into 1 ml freezing tubes. A representative aliquot is freeze thawed and titred to determine virus concentration. Virus stocks are stored below −20° C.
  • Semi-Pure Preparations [0250]
  • Infected cells are harvested as described previously (Earl et al a & b; Earl and Moss; 1991). After centrifugation cells are resuspended in PBS (2 ml/175 cm[0251] 2 flask) and homogenised by 30-40 strokes in a tight fitting glass dounce homogeniser, on ice. Cell breakage is checked by microscopy. Nuclei, cellular organelles and membranes are removed by a centrifugation at 300 g for 5 mins (4° C.), keep supernatant. The cell pellet is resuspended in 1 ml/175 cm2 flask and centrifugation repeated. The supernatants are pooled, aliquoted and stored.
  • Purified Preparation [0252]
  • Infected cells are harvested as previously described (Earl et al.a & b; Earl and Moss; 1991) and resuspended in 10 mM Tris.Cl, pH 9.0 (2 ml/flask), keeping samples on ice from this point of the procedure. Homogenise as described previously using 10 mM Tris. The lysate is sonicated (on ice) using an XL 2015 sonicating cup (Misonics, USA) at maximum output (500 W) for 1 min. The sample is placed on ice for 1 min and the sonication repeated up to 3 times. A maximum of 5 ml is sonicated at a time, and ice is replenished during sonication. The lysate is gently layered onto a cushion of 17 ml of 36% sucrose (in 10 mM Tris.Cl, pH 9.0) in a SW-27 centrifuge tube. Lyates are centrifuged for 80 mins in an SW-27 rotor at 13 500 rpm (32,900×g), 4° C. The supernatant is discarded and the viral pellet resuspended in sterile PBS and sonicated in a cup sonicator for 1 min (on ice). Concentrated virus is aliquoted and stored at below −20° C. [0253]
  • EXAMPLE 22 Production of EIAV Vector Particles from MVA-EIAV Hybrids
  • As described above large scale preparations of recombinant MVA-EIAV are made. These preparations are used to infect mammalian cells that are non-permissive for MVA, such that the resulting supernatant will only contain EIAV and not infectious MVA (Meyer et al 1991, Carroll and Moss 1997). A suitable cell line is MRC5 (ATCC). Cells are infected at an MOI of 3. Infections are allowed to run for approximately 48 hours before supernatants are harvested and EIAV vector particles either used directly or concentrated/purified by ultracentrifugation or cross-flow methods. To produce large scale preparations, are grown in suspension or on microcarriers or in roller bottles. EIAV vectors carrying gene of interest prepared in these ways are used to transduce target cells in vivo or in vitro. [0254]
  • REFERENCES
  • Blomer, U., Naldini, L., Kafri, T., Trono, D., Verma, I. M., and Gage, F. H. (1997). Highly efficient and sustained gene transfer in adult neurons with a lentivirus vector. J Virol 71, 6641-6649. [0255]
  • Blomer, U., Naldini, L., Verma, I. M., Trono, D., and Gage, F. H. (1996). Applications of gene therapy to the CNS. [0256] Hum Mol Genet 5 Spec No, 1397-404.
  • Clever, J., Sassetti, C., and Parslow, T. G. (1995). RNA secondary structure and binding sites for gag gene products in the 5′ packaging signal of human [0257] immunodeficiency virus type 1. J Virol 69, 2101-9.
  • Clever, J. L., and Parslow, T. G. (1997). Mutant human [0258] immunodeficiency virus type 1 genomes with defects in RNA dimerization or encapsidation. J Virol 71, 3407-14.
  • Fields, B. N., Knipe, D. M., and Howley, P. M. (1996). Fields Virology, R. M. Chanock, J. L. Melnick, T. P. Monath, B. Roizman and S. E. Straus, eds. (Philadelphia. New York: Lippincott—Raven Publishers). [0259]
  • Fuller S, von Bonsdorff CH, Simons K. Vesicular stomatitis virus infects and matures only through the basolateral surface of the polarized epithelial cell line, MDCK. Cell 1984 Aug;38(1):65-77 [0260]
  • Harrison, G. S., Long, C. J., Maxwell, F., Glode, L. M., and Maxwell, I. H. (1992). Inhibition of HIV production in cells containing an integrated, HIV-regulated diphtheria toxin A chain Gene. AIDs Research and [0261] Human Retrovirus 8, 39-45.
  • Hayashi T, Shioda T, Iwakura Y, Shibuta H. RNA packaging signal of human [0262] immunodeficiency virus type 1. Virology 1992 June;188(2):590-599
  • Kim V. N., Mitrophanous K., Kingsman S. M., Kingsman A. J. 1998. Minimal Requirement for a Lentiviral Vector Based on Human [0263] Immunodeficiency Virus Type 1. J. Virol. 1998 72:811-6.
  • Kim, S. Y., R. Byrn, J. Groopman, and D. Baltimore. 1989. Temporal aspects of DNA and RNA synthesis during human immunodeficiency virus infection: evidence for differential gene expression. J. Virol. 63:3708-3713. [0264]
  • Lewis, P. F., and M. Emerman. 1994. Passage through mitosis is required for oncoretroviruses but-not for the human immunodeficiency virus. J Virol. 68:510-6. [0265]
  • Mann R, Mulligan RC, Baltimore D. Construction of a retrovirus packaging mutant and its use to produce helper-free defective retrovirus. Cell 1983 May;33(1):153-159 [0266]
  • Martarano, L., Stephens, R., Rice, N., and Derse, D. (1994). Equine infectious anemia virus trans-regulatory protein Rev controls viral mRNA stability, accumulation, and alternative splicing. J Virol 68, 3102-11. [0267]
  • Naldini, L., Blomer, U., Gage, F. H., Trono, D., and Verma, I. M. (1996). Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. Proc Natl Acad Sci U S A 93, 11382-11388. [0268]
  • Naldini, L., Blomer, U., Gallay, P., Ory, D., Mulligan, R., Gage, F. H., Verma, I. M., and Trono, D (1996). In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector [see comments]. Science 272, 263-7. [0269]
  • Payne, S. L., Rausch, J., Rushlow, K., Montelaro, R. C., Issel, C., Flaherty, M., Perry, S., Sellon, D., and Fuller, F. (1994). Characterization of infectious molecular clones of equine infectious anaemia virus. J Gen Virol 75, 425-9. [0270]
  • Yee, J.-K., M. Atsushi, P. LaPorte, K. Bouic, J. C. Bums, and T. Friedmann (1994) A general method for th generation of high-titer, pantropic retroviral vectors: Highly efficient infection of primary hepatocytes. Proc. Natl. Acad. Sci. USA 91:9564-9568. [0271]
  • Zufferey, R., Nagy, D., Mandel, R. J., Naldini, L., and Trono, D. (1997). Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo. [0272] Nat Biotechnol 15, 871-875.
  • Cannon 1996 J Virol 1996 70:8234-40. Murine leukemia virus-based Tat-inducible long terminal repeat replacement vectors: a new system for anti-human immunodeficiency virus gene therapy. Cannon P M, Kim N, Kingsman S M, Kingsman A J. [0273]
  • Carroll M W, Moss B [0274] E. coli beta-glucuronidase (GUS) as a marker for recombinant vaccinia viruses. Biotechniques 1995 19:352-4
  • Carroll MW, Moss B Host range and cytopathogenicity of the highly attenuated MVA strain of vaccinia virus: propagation and generation of recombinant viruses in a non-human mammalian cell line. Virology 1997 238:198-211 [0275]
  • Chakrabarti S, Brechling K, Moss B Vaccinia virus expression vector: coexpression of beta-galactosidase provides visual screening of recombinant virus plaques. Mol Cell Biol 1985 12:3403-9 [0276]
  • Chakrabarti S, Sisler J R, Moss B Biotechniques 1997 6:1094-7 Compact, synthetic, vaccinia virus early/late promoter for protein expression. Davison 1989a J Mol Biol 1989 20;210(4):749-69. Structure of vaccinia virus early promoters. Davison A J, Moss B [0277]
  • Davison 1989b J Mol Biol 1989 210(4):771-84. Structure of vaccinia virus late promoters. Davison A J, Moss B [0278]
  • P L. Earl (a), N. Cooper, L S Wyatt, B. Moss & M. W. Carroll 1998 Preparation of Cell Cultures and Vaccinia Virus Stocks. Current Protocols in Molecular Biology Supplement 43 Unit 16.16. John Wiley and Sons Inc. [0279]
  • P L. Earl (b), B. Moss L. S. Wyatt, & M. W. Carroll 1998 Generation of Recombinant Vaccinia Viruses. Current Protocols in Molecular Biology. Supplement 43 Unit 16.17. Current Protocols in Molecular Biology. John Wiley and Sons Inc. [0280]
  • Flexner C, Hugin A, Moss B Nature 1987 330(6145):259-62 Prevention of vaccinia virus infection in immunodeficient mice by vector-directed IL-2 expression. [0281]
  • Holzer G W, Falkner F G Construction of a vaccinia virus deficient in the essential DNA repair enzyme uracil DNA glycosylase by a complementing cell line. J Virol 1997 71:4997-5002 [0282]
  • Kim V N, Mitrophanous K, Kingsman S M, Kingsman A J Minimal requirement for a lentivirus vector based on human [0283] immunodeficiency virus type 1. J Virol 1998 72:811-6
  • Mackett M, Smith G L, Moss B Vaccinia virus: a selectable eukaryotic cloning and expression vector. Proc Natl Acad Sci U S A 1982 7923:7415-9 [0284]
  • Mahnel H, Mayr A Berl Munch Tierarztl Wochenschr 1994 Aug;107(8):253-6 [Experiences with immunization against orthopox viruses of humans and animals using vaccine strain MVA].[Article in German] Zentralbl Bakteriol [B] 1978 Dec; 167(5-6):375-90 [0285]
  • Martarano L, Stephens R, Rice N, Derse D Equine infectious anemia virus trans-regulatory protein Rev controls viral mRNA stability, accumulation, and alternative splicing. J Virol 1994 May;68(5):3102-11 [0286]
  • Mayr A, Stickl H, Muller H K, Danner K, Singer H [The smallpox vaccination strain MVA: marker, genetic structure, experience gained with the parenteral vaccination and behavior in organisms with a debilitated defence mechanism]. Zentralbl Bakteriol [B]. 1978 Dec;167(5-6):375-90. German. [0287]
  • Meyer H, Sutter G, Mayr A Mapping of deletions in the genome of the highly attenuated vaccinia virus MVA and their influence on virulence. J Gen Virol 1991 72:1031-8 [0288]
  • Moss B Poxviridae: The viruses and their replication Chapter 83. p2637-2672. In Fields, B. N., Knipe, D. M. & Howley, P.M. Fields Virology. Third Edition edn Vol. 2 eds Chanock, R. M., Melnick, J. L., Monath, T. P., Roizman, B. & Straus, S. E. Lippincott—Raven Publishers, Philadelphia. New York, 1996 [0289]
  • Moss B, Carroll M W, Wyatt L S, Bennink J R, Hirsch V M, Goldstein S, Elkins W R, Fuerst T R, Lifson J D, Piatak M, Restifo N P, Overwijk W, Chamberlain R, Rosenberg S A, Sutter G Host range restricted, non-replicating vaccinia virus vectors as vaccine candidates. Adv Exp Med Biol 1996;397:7-13 [0290]
  • Panicali D, Paoletti E Construction of poxviruses as cloning vectors: insertion of the thymidine kinase gene from herpes simplex virus into the DNA of infectious vaccinia virus. Proc Natl Acad Sci U S A 1982 79:4927-31 [0291]
  • Soneoka Y, Cannon P M, Ramsdale E E, Griffiths J C, Romano G, Kingsman SM, Kingsman A J 1995 Nucleic Acids Res 1995 23628-33. A transient three-plasmid expression system for the production of high titer retroviral vectors. [0292]
  • Sutter G, Moss B Nonreplicating vaccinia vector efficiently expresses recombinant genes. Proc Natl Acad Sci U S A Nov. 15, 1992;89(22):10847-51 [0293]
  • Taylor J, Weinberg R, Tartaglia J, Richardson C, Alkhatib G, Briedis D, Appel M, Norton E, Paoletti E Nonreplicating viral vectors as potential vaccines: recombinant canarypox virus expressing measles virus fusion (F) and hemagglutinin (HA) glycoproteins. Virology 1992 Mar;187(1):321-8 [0294]
  • Paoletti E, Tartaglia J, Taylor J Safe and effective poxvirus vectors—NYVAC and ALVAC. Dev Biol Stand 1994;82:65-9 [0295]
  • Wyatt L S, Moss B, Rozenblatt S Replication-deficient vaccinia virus encoding bacteriophage T7 RNA polymerase for transient gene expression in mammalian cells. Virology Jun. 20, 1995;210(1):202-5 [0296]
  • Wyatt L S, Carroll M W, Czerny C P, Merchlinsky M, Sisler J R, Moss B Marker rescue of the host range restriction defects of modified vaccinia virus Ankara. Virology 1998 251:334-42 [0297]
  • Wyatt L S, Shors S T, Murphy B R, Moss B Development of a replication-deficient recombinant vaccinia virus vaccine effective against [0298] parainfluenza virus 3 infection in an animal model. Vaccine 1996 Oct; 14(15):1451-8
  • 1 64 1 381 RNA Equine infectious anemia virus 1 augauaccgg gcacucagau ucugcggucu gagucccuuc ucugcugggc ugaaaaggcc 60 uuuguauaaa uauaauucuc uacucagucc cugucucuag uuugucuguu cgagauccua 120 caguuggcgc ccgaacaggg accugagggg gcgcagaccc uaccuguuga accuggcuga 180 ucguaggauc cccgggacag cagaggagaa cuuacagaag ucuucuggag guguuccugg 240 ggagaacaca ggaggacagg uaagauggga gacccuuuga cauggagcaa ggcgcucaag 300 aaguuaagaa ggugacggua caagggucuc aguuaacucu gguaacugua auugggcgcu 360 aagucuaggu agacuuauuu c 381 2 41 DNA Artificial Sequence misc_feature (1)..(41) sequence showing part of split polyA signal 2 tcgctgcagc ggaataaagg gcaggtaagt atcaaggtta c 41 3 60 DNA Artificial Sequence, primer misc_feature (1)..(60) sequence showing the part of split polyA signal 3 tcgctgcagc ggacacacaa aaaaccaaca cacagaactg ggaagtggac acctgtggag 60 4 63 DNA Artificial Sequence misc_feature (1)..(63) sequence showing both the parts of polyA signal 4 aataaagggc aggtaagctc cacaggtgtc cactccagtt ctgtgtgttg gttttttgtg 60 tgt 63 5 50 DNA Artificial Sequence polyA_signal (1)..(50) sequence of the polyA signal 5 aataaagggc aggtgtccac tccagttctg tgtgttggtt ttttgtgtgt 50 6 33 DNA Artificial Sequence, primer misc_feature (1)..(33) primer 6 tcgatagatc tgagtccgtt acataactta cgg 33 7 57 DNA Artificial Sequence,primer misc_feature (1)..(57) primer 7 gatctcgaac agacaaacta gagacaggga ctgcaaacag caagaggctt tattggg 57 8 30 DNA Artificial Sequence,primer misc_feature (1)..(30) primer 8 gtccctgtct ctagtttgtc tgttcgagat 30 9 27 DNA Artificial Sequence,primer misc_feature (1)..(27) primer 9 ggggatccac tagttctaga gatattc 27 10 27 DNA Artificial Sequence,primer misc_feature (1)..(27) primer 10 ccttagacct ggagattcga agcgaag 27 11 53 DNA Artificial Sequence,primer misc_feature (1)..(53) primer 11 ccaaacctac aggtggggtc tttcatttac aaggttatga gagcatcagc aac 53 12 27 DNA Artificial Sequence,primer misc_feature (1)..(27) primer 12 aatgaaagac cccacctgta ggtttgg 27 13 41 DNA Artificial Sequence,primer misc_feature (1)..(41) primer 13 gtagagtgcc caattgccag tatacactcc gctatcgcta c 41 14 11299 DNA Artificial Sequence misc_feature (1)..(11299) plasmid 14 ctaaattgta agcgttaata ttttgttaaa attcgcgtta aatttttgtt aaatcagctc 60 attttttaac caataggccg aaatcggcaa aatcccttat aaatcaaaag aatagaccga 120 gatagggttg agtgttgttc cagtttggaa caagagtcca ctattaaaga acgtggactc 180 caacgtcaaa gggcgaaaaa ccgtctatca gggcgatggc ccactacgtg aaccatcacc 240 ctaatcaagt tttttggggt cgaggtgccg taaagcacta aatcggaacc ctaaagggag 300 cccccgattt agagcttgac ggggaaagcc aacctggctt atcgaaatta atacgactca 360 ctatagggag accggcagat ctgagtccgt tacataactt acggtaaatg gcccgcctgg 420 ctgaccgccc aacgaccccc gcccattgac gtcaataatg acgtatgttc ccatagtaac 480 gccaataggg actttccatt gacgtcaatg ggtggagtat ttacggtaaa ctgcccactt 540 ggcagtacat caagtgtatc atatgccaag tacgccccct attgacgtca atgacggtaa 600 atggcccgcc tggcattatg cccagtacat gaccttatgg gactttccta cttggcagta 660 catctacgta ttagtcatcg ctattaccat ggtgatgcgg ttttggcagt acatcaatgg 720 gcgtggatag cggtttgact cacggggatt tccaagtctc caccccattg acgtcaatgg 780 gagtttgttt tggcaccaaa atcaacggga ctttccaaaa tgtcgtaaca actccgcccc 840 attgacgcaa atgggcggta ggcgtgtacg gtgggaggtc tatataagca gagctcgttt 900 agtgaaccgc gccagtcttc cgatagactg cgtcgcccgg gtacccgtat tcccaataaa 960 gcctcttgct gtttgcatcc gaatcgtggt ctcgctgttc cttgggaggg tctcctctga 1020 gtgattgact acccacgacg ggggtctttc atttctctag tttgtctgtt cgagatccta 1080 cagttggcgc ccgaacaggg acctgagagg ggcgcagacc ctacctgttg aacctggctg 1140 atcgtaggat ccccgggaca gcagaggaga acttacagaa gtcttctgga ggtgttcctg 1200 gccagaacac aggaggacag gtaagatggg agaccctttg acatggagca aggcgctcaa 1260 gaagttagag aaggtgacgg tacaagggtc tcagaaatta actactggta actgtaattg 1320 ggcgctaagt ctagtagact tatttcatga taccaacttt gtaaaagaaa aggactggca 1380 gctgagggat gtcattccat tgctggaaga tgtaactcag acgctgtcag gacaagaaag 1440 agaggccttt gaaagaacat ggtgggcaat ttctgctgta aagatgggcc tccagattaa 1500 taatgtagta gatggaaagg catcattcca gctcctaaga gcgaaatatg aaaagaagac 1560 tgctaataaa aagcagtctg agccctctga agaatatctc tagagtgtga ttttaagggc 1620 gaattctgca ggagtgggga ggcacgatgg ccgctttggt cgaggcggat ccggccatta 1680 gccatattat tcattggtta tatagcataa atcaatattg gctattggcc attgcatacg 1740 ttgtatccat atcataatat gtacatttat attggctcat gtccaacatt accgccatgt 1800 tgacattgat tattgactag ttattaatag taatcaatta cggggtcatt agttcatagc 1860 ccatatatgg agttccgcgt tacataactt acggtaaatg gcccgcctgg ctgaccgccc 1920 aacgaccccc gcccattgac gtcaataatg acgtatgttc ccatagtaac gccaataggg 1980 actttccatt gacgtcaatg ggtggagtat ttacggtaaa ctgcccactt ggcagtacat 2040 caagtgtatc atatgccaag tacgccccct attgacgtca atgacggtaa atggcccgcc 2100 tggcattatg cccagtacat gaccttatgg gactttccta cttggcagta catctacgta 2160 ttagtcatcg ctattaccat ggtgatgcgg ttttggcagt acatcaatgg gcgtggatag 2220 cggtttgact cacggggatt tccaagtctc caccccattg acgtcaatgg gagtttgttt 2280 tggcaccaaa atcaacggga ctttccaaaa tgtcgtaaca actccgcccc attgacgcaa 2340 atgggcggta ggcatgtacg gtgggaggtc tatataagca gagctcgttt agtgaaccgt 2400 cagatcgcct ggagacgcca tccacgctgt tttgacctcc atagaagaca ccgggaccga 2460 tccagcctcc gcggccccaa gcttcagctg ctcgaggatc tgcggatccg gggaattccc 2520 cagtctcagg atccaccatg ggggatcccg tcgttttaca acgtcgtgac tgggaaaacc 2580 ctggcgttac ccaacttaat cgccttgcag cacatccccc tttcgccagc tggcgtaata 2640 gcgaagaggc ccgcaccgat cgcccttccc aacagttgcg cagcctgaat ggcgaatggc 2700 gctttgcctg gtttccggca ccagaagcgg tgccggaaag ctggctggag tgcgatcttc 2760 ctgaggccga tactgtcgtc gtcccctcaa actggcagat gcacggttac gatgcgccca 2820 tctacaccaa cgtaacctat cccattacgg tcaatccgcc gtttgttccc acggagaatc 2880 cgacgggttg ttactcgctc acatttaatg ttgatgaaag ctggctacag gaaggccaga 2940 cgcgaattat ttttgatggc gttaactcgg cgtttcatct gtggtgcaac gggcgctggg 3000 tcggttacgg ccaggacagt cgtttgccgt ctgaatttga cctgagcgca tttttacgcg 3060 ccggagaaaa ccgcctcgcg gtgatggtgc tgcgttggag tgacggcagt tatctggaag 3120 atcaggatat gtggcggatg agcggcattt tccgtgacgt ctcgttgctg cataaaccga 3180 ctacacaaat cagcgatttc catgttgcca ctcgctttaa tgatgatttc agccgcgctg 3240 tactggaggc tgaagttcag atgtgcggcg agttgcgtga ctacctacgg gtaacagttt 3300 ctttatggca gggtgaaacg caggtcgcca gcggcaccgc gcctttcggc ggtgaaatta 3360 tcgatgagcg tggtggttat gccgatcgcg tcacactacg tctgaacgtc gaaaacccga 3420 aactgtggag cgccgaaatc ccgaatctct atcgtgcggt ggttgaactg cacaccgccg 3480 acggcacgct gattgaagca gaagcctgcg atgtcggttt ccgcgaggtg cggattgaaa 3540 atggtctgct gctgctgaac ggcaagccgt tgctgattcg aggcgttaac cgtcacgagc 3600 atcatcctct gcatggtcag gtcatggatg agcagacgat ggtgcaggat atcctgctga 3660 tgaagcagaa caactttaac gccgtgcgct gttcgcatta tccgaaccat ccgctgtggt 3720 acacgctgtg cgaccgctac ggcctgtatg tggtggatga agccaatatt gaaacccacg 3780 gcatggtgcc aatgaatcgt ctgaccgatg atccgcgctg gctaccggcg atgagcgaac 3840 gcgtaacgcg aatggtgcag cgcgatcgta atcacccgag tgtgatcatc tggtcgctgg 3900 ggaatgaatc aggccacggc gctaatcacg acgcgctgta tcgctggatc aaatctgtcg 3960 atccttcccg cccggtgcag tatgaaggcg gcggagccga caccacggcc accgatatta 4020 tttgcccgat gtacgcgcgc gtggatgaag accagccctt cccggctgtg ccgaaatggt 4080 ccatcaaaaa atggctttcg ctacctggag agacgcgccc gctgatcctt tgcgaatacg 4140 cccacgcgat gggtaacagt cttggcggtt tcgctaaata ctggcaggcg tttcgtcagt 4200 atccccgttt acagggcggc ttcgtctggg actgggtgga tcagtcgctg attaaatatg 4260 atgaaaacgg caacccgtgg tcggcttacg gcggtgattt tggcgatacg ccgaacgatc 4320 gccagttctg tatgaacggt ctggtctttg ccgaccgcac gccgcatcca gcgctgacgg 4380 aagcaaaaca ccagcagcag tttttccagt tccgtttatc cgggcaaacc atcgaagtga 4440 ccagcgaata cctgttccgt catagcgata acgagctcct gcactggatg gtggcgctgg 4500 atggtaagcc gctggcaagc ggtgaagtgc ctctggatgt cgctccacaa ggtaaacagt 4560 tgattgaact gcctgaacta ccgcagccgg agagcgccgg gcaactctgg ctcacagtac 4620 gcgtagtgca accgaacgcg accgcatggt cagaagccgg gcacatcagc gcctggcagc 4680 agtggcgtct ggcggaaaac ctcagtgtga cgctccccgc cgcgtcccac gccatcccgc 4740 atctgaccac cagcgaaatg gatttttgca tcgagctggg taataagcgt tggcaattta 4800 accgccagtc aggctttctt tcacagatgt ggattggcga taaaaaacaa ctgctgacgc 4860 cgctgcgcga tcagttcacc cgtgcaccgc tggataacga cattggcgta agtgaagcga 4920 cccgcattga ccctaacgcc tgggtcgaac gctggaaggc ggcgggccat taccaggccg 4980 aagcagcgtt gttgcagtgc acggcagata cacttgctga tgcggtgctg attacgaccg 5040 ctcacgcgtg gcagcatcag gggaaaacct tatttatcag ccggaaaacc taccggattg 5100 atggtagtgg tcaaatggcg attaccgttg atgttgaagt ggcgagcgat acaccgcatc 5160 cggcgcggat tggcctgaac tgccagctgg cgcaggtagc agagcgggta aactggctcg 5220 gattagggcc gcaagaaaac tatcccgacc gccttactgc cgcctgtttt gaccgctggg 5280 atctgccatt gtcagacatg tataccccgt acgtcttccc gagcgaaaac ggtctgcgct 5340 gcgggacgcg cgaattgaat tatggcccac accagtggcg cggcgacttc cagttcaaca 5400 tcagccgcta cagtcaacag caactgatgg aaaccagcca tcgccatctg ctgcacgcgg 5460 aagaaggcac atggctgaat atcgacggtt tccatatggg gattggtggc gacgactcct 5520 ggagcccgtc agtatcggcg gaattccagc tgagcgccgg tcgctaccat taccagttgg 5580 tctggtgtca aaaataataa taaccgggca ggggggatcc gcagatccgg ctgtggaatg 5640 tgtgtcagtt agggtgtgga aagtccccag gctccccagc aggcagaagt atgcaaagca 5700 tgcctgcagg aattcgatat caagcttatc gataccgtcg acctcgaggg ggggcccggt 5760 acccagcttt tgttcccttt agtgagggtt aattgcgcgg gaagtattta tcactaatca 5820 agcacaagta atacatgaga aacttttact acagcaagca caatcctcca aaaaattttg 5880 tttttacaaa atccctggtg aacatgattg gaagggacct actagggtgc tgtggaaggg 5940 tgatggtgca gtagtagtta atgatgaagg aaagggaata attgctgtac cattaaccag 6000 gactaagtta ctaataaaac caaattgagt attgttgcag gaagcaagac ccaactacca 6060 ttgtcagctg tgtttcctga ggtctctagg aattgattac ctcgatgctt cattaaggaa 6120 gaagaataaa caaagactga aggcaatcca acaaggaaga caacctcaat atttgttata 6180 aggtttgata tatgggagta tttggtaaag gggtaacatg gtcagcatcg cattctatgg 6240 gggaatccca gggggaatct caacccctat tacccaacag tcagaaaaat ctaagtgtga 6300 ggagaacaca atgtttcaac cttattgtta taataatgac agtaagaaca gcatggcaga 6360 atcgaaggaa gcaagagacc aagaaatgaa cctgaaagaa gaatctaaag aagaaaaaag 6420 aagaaatgac tggtggaaaa taggtatgtt tctgttatgc ttagcaggaa ctactggagg 6480 aatactttgg tggtatgaag gactcccaca gcaacattat atagggttgg tggcgatagg 6540 gggaagatta aacggatctg gccaatcaaa tgctatagaa tgctggggtt ccttcccggg 6600 gtgtagacca tttcaaaatt acttcagtta tgagaccaat agaagcatgc atatggataa 6660 taatactgct acattattag aagctttaac caatataact gctctataaa taacaaaaca 6720 gaattagaaa catggaagtt agtaaagact tctggcataa ctcctttacc tatttcttct 6780 gaagctaaca ctggactaat tagacataag agagattttg gtataagtgc aatagtggca 6840 gctattgtag ccgctactgc tattgctgct agcgctacta tgtcttatgt tgctctaact 6900 gaggttaaca aaataatgga agtacaaaat catacttttg aggtagaaaa tagtactcta 6960 aatggtatgg atttaataga acgacaaata aagatattat atgctatgat tcttcaaaca 7020 catgcagatg ttcaactgtt aaaggaaaga caacaggtag aggagacatt taatttaatt 7080 ggatgtatag aaagaacaca tgtattttgt catactggtc atccctggaa tatgtcatgg 7140 ggacatttaa atgagtcaac acaatgggat gactgggtaa gcaaaatgga agatttaaat 7200 caagagatac taactacact tcatggagcc aggaacaatt tggcacaatc catgataaca 7260 ttcaatacac cagatagtat agctcaattt ggaaaagacc tttggagtca tattggaaat 7320 tggattcctg gattgggagc ttccattata aaatatatag tgatgttttt gcttatttat 7380 ttgttactaa cctcttcgcc taagatcctc agggccctct ggaaggtgac cagtggtgca 7440 gggtcctccg gcagtcgtta cctgaagaaa aaattccatc acaaacatgc atcgcgagaa 7500 gacacctggg accaggccca acacaacata cacctagcag gcgtgaccgg tggatcaggg 7560 gacaaatact acaagcagaa gtactccagg aacgactgga atggagaatc agaggagtac 7620 aacaggcggc caaagagctg ggtgaagtca atcgaggcat ttggagagag ctatatttcc 7680 gagaagacca aaggggagat ttctcagcct ggggcggcta tcaacgagca caagaacggc 7740 tctgggggga acaatcctca ccaagggtcc ttagacctgg agattcgaag cgaaggagga 7800 aacatttatg actgttgcat taaagcccaa gaaggaactc tcgctatccc ttgctgtgga 7860 tttcccttat ggctattttg gggactagta attatagtag gacgcatagc aggctatgga 7920 ttacgtggac tcgctgttat aataaggatt tgtattagag gcttaaattt gatatttgaa 7980 ataatcagaa aaatgcttga ttatattgga agagctttaa atcctggcac atctcatgta 8040 tcaatgcctc agtatgttta gaaaaacaag gggggaactg tggggttttt atgaggggtt 8100 ttataaatga ttataagagt aaaaagaaag ttgctgatgc tctcataacc ttgtaaatga 8160 aagaccccac ctgtaggttt ggcaagctag cttaagtaac gccattttgc aaggcatgga 8220 aaaatacata actgagaata gagaagttca gatcaaggtc aggaacagat ggaacagctg 8280 aatatgggcc aaacaggata tctgtggtaa gcagttcctg ccccggctca gggccaagaa 8340 cagatggaac agctgaatat gggccaaaca ggatatctgt ggtaagcagt tcctgccccg 8400 gctcagggcc aagaacagat ggtccccaga tgcggtccag ccctcagcag tttctagaga 8460 accatcagat gtttccaggg tgccccaagg acctgaaatg accctgtgcc ttatttgaac 8520 taaccaatca gttcgcttct cgcttctgtt cgcgcgcttc tgctccccga gctcaataaa 8580 agagcccaca acccctcact cggggcgcca gtcctccgat tgactgagtc gcccgggtac 8640 ccgtgtatcc aataaaccct cttgcagttg catccgactt gtggtctcgc tgttccttgg 8700 gagggtctcc tctgagtgat tgactacccg tcagcggggg tctttcattt gggggctcgt 8760 ccgggatcgg gagacccctg cccagggacc accgacccac caccgggagg taagctggct 8820 gcctcgcgcg tttcggtgat gacggtgaaa acctctgaca catgcagctc ccggagacgg 8880 tcacagcttg tctgtaagcg gatgccggga gcagacaagc ccgtcagggc gcgtcagcgg 8940 gtgttggcgg gtgtcggggc gcagccatga cccagtcacg tagcgatagc ggagtgtata 9000 ctggcaattg ggcactcaga ttctgcggtc tgagtccctt ctctgctggg ctgaaaaggc 9060 ctttgtaata aatataattc tctactcagt ccctgtctct agtttgtctg ttcgagatcc 9120 tacagagctc atgccttggc gtaatcatgg tcatagctgt ttcctgtgtg aaattgttat 9180 ccgctcacaa ttccacacaa catacgagcc ggaagcataa agtgtaaagc ctggggtgcc 9240 taatgagtga gctaactcac attaattgcg ttgcgctcac tgcccgcttt ccagtcggga 9300 aacctgtcgt gccagctgca ttaatgaatc ggccaacgcg cggggagagg cggtttgcgt 9360 attgggcgct cttccgcttc ctcgctcact gactcgctgc gctcggtcgt tcggctgcgg 9420 cgagcggtat cagctcactc aaaggcggta atacggttat ccacagaatc aggggataac 9480 gcaggaaaga acatgtgagc aaaaggccag caaaaggcca ggaaccgtaa aaaggccgcg 9540 ttgctggcgt ttttccatag gctccgcccc cctgacgagc atcacaaaaa tcgacgctca 9600 agtcagaggt ggcgaaaccc gacaggacta taaagatacc aggcgtttcc ccctggaagc 9660 tccctcgtgc gctctcctgt tccgaccctg ccgcttaccg gatacctgtc cgcctttctc 9720 ccttcgggaa gcgtggcgct ttctcatagc tcacgctgta ggtatctcag ttcggtgtag 9780 gtcgttcgct ccaagctggg ctgtgtgcac gaaccccccg ttcagcccga ccgctgcgcc 9840 ttatccggta actatcgtct tgagtccaac ccggtaagac acgacttatc gccactggca 9900 gcagccactg gtaacaggat tagcagagcg aggtatgtag gcggtgctac agagttcttg 9960 aagtggtggc ctaactacgg ctacactaga aggacagtat ttggtatctg cgctctgctg 10020 aagccagtta ccttcggaaa aagagttggt agctcttgat ccggcaaaca aaccaccgct 10080 ggtagcggtg gtttttttgt ttgcaagcag cagattacgc gcagaaaaaa aggatctcaa 10140 gaagatcctt tgatcttttc tacggggtct gacgctcagt ggaacgaaaa ctcacgttaa 10200 gggattttgg tcatgagatt atcaaaaagg atcttcacct agatcctttt aaattaaaaa 10260 tgaagtttta aatcaatcta aagtatatat gagtaaactt ggtctgacag ttaccaatgc 10320 ttaatcagtg aggcacctat ctcagcgatc tgtctatttc gttcatccat agttgcctga 10380 ctccccgtcg tgtagataac tacgatacgg gagggcttac catctggccc cagtgctgca 10440 atgataccgc gagacccacg ctcaccggct ccagatttat cagcaataaa ccagccagcc 10500 ggaagggccg agcgcagaag tggtcctgca actttatccg cctccatcca gtctattaat 10560 tgttgccggg aagctagagt aagtagttcg ccagttaata gtttgcgcaa cgttgttgcc 10620 attgctacag gcatcgtggt gtcacgctcg tcgtttggta tggcttcatt cagctccggt 10680 tcccaacgat caaggcgagt tacatgatcc cccatgttgt gcaaaaaagc ggttagctcc 10740 ttcggtcctc cgatcgttgt cagaagtaag ttggccgcag tgttatcact catggttatg 10800 gcagcactgc ataattctct tactgtcatg ccatccgtaa gatgcttttc tgtgactggt 10860 gagtactcaa ccaagtcatt ctgagaatag tgtatgcggc gaccgagttg ctcttgcccg 10920 gcgtcaatac gggataatac cgcgccacat agcagaactt taaaagtgct catcattgga 10980 aaacgttctt cggggcgaaa actctcaagg atcttaccgc tgttgagatc cagttcgatg 11040 taacccactc gtgcacccaa ctgatcttca gcatctttta ctttcaccag cgtttctggg 11100 tgagcaaaaa caggaaggca aaatgccgca aaaaagggaa taagggcgac acggaaatgt 11160 tgaatactca tactcttcct ttttcaatat tattgaagca tttatcaggg ttattgtctc 11220 atgagcggat acatatttga atgtatttag aaaaataaac aaataggggt tccgcgcaca 11280 tttccccgaa aagtgccac 11299 15 66 DNA Artificial Sequence,primer misc_feature (1)..(66) primer 15 atcgaagctt aattaaaagt agaaaatata ttctaattta ttgggcactc agttctgcgg 60 tctgag 66 16 35 DNA Artificial Sequence,primer misc_feature (1)..(35) primer 16 tcagctgcag ttcgggcgcc aactgtagga tctcg 35 17 33 DNA Artificial Sequence,primer misc_feature (1)..(33) primer 17 actgctgcag agattcgaag cgaaggagga aac 33 18 31 DNA Artificial Sequence,primer misc_feature (1)..(31) primer 18 tgtggggttt ccatgagggg ttttataaat g 31 19 30 DNA Artificial Sequence,primer misc_feature (1)..(30) primer 19 ccctcatgga aaccccacgt tccccccttg 30 20 33 DNA Artificial Sequence,primer misc_feature (1)..(33) primer 20 ctgaagatct gaatctgagt gcccaattgt cag 33 21 23 DNA Artificial Sequence,primer misc_feature (1)..(23) primer 21 ctgacaattg ggcactcaga ttc 23 22 44 DNA Artificial Sequence,primer misc_feature (1)..(44) primer 22 catgagatct taaaaaaaaa tgatgagaga attatattta ttac 44 23 21 DNA Equine infectious anemia virus misc_feature (1)..(21) 23 gggcactcag attctgcggt c 21 24 77 DNA Equine infectious anemia virus 24 cuagugauuc ugagugcccc ugaugagcgg ccgaaaggcc gcgaaaccug cguacgacac 60 gcaggucggg cactcag 77 25 50 DNA Artificial Sequence promoter (13)..(29) T7 promoter 25 atcgttaatt aataatacga ctcactatag ggcactcaga ttctgcggtc 50 26 82 DNA Artificial Sequence terminator (11)..(59) T7 termination sequence 26 catgagatct caaaaaaccc ctcaagaccc gtttagaggc cccaaggggt tatgctagtg 60 atgagagaat tatatttatt ac 82 27 7252 DNA Artificial Sequence, plasmid misc_feature (1)..(7252) plasmid vector 27 agcttttgcg atcaataaat ggatcacaac cagtatctct taacgatgtt cttcgcagat 60 gatgattcat tttttaagta tttggctagt caagatgatg aaatcttcat tatctgatat 120 attgcaaatc actcaatatc tagactttct gttattatta ttgatccaat caaaaaataa 180 attagaagcc gtgggtcatt gttatgaatc tctttcagag gaatacagac aattgacaaa 240 attcacagac tttcaagatt ttaaaaaact gtttaacaag gtccctattg ttacagatgg 300 aagggtcaaa cttaataaag gatatttgtt cgactttgtg attagtttga tgcgattcaa 360 aaaagaatcc tctctagcta ccaccgcaat agatcctgtt agatacatag atcctcgtcg 420 caatatcgca ttttctaacg tgatggatat attaaagtcg aataaagtga acaataatta 480 attctttatt gtcatcatga acggcggaca tattcagttg ataatcggcc ccatgttttc 540 aggtaaaagt acagaattaa ttagacgagt tagacgttat caaatagctc aatataaatg 600 cgtgactata aaatattcta acgataatag atacggaacg ggactatgga cgcatgataa 660 gaataatttt gaagcattgg aagcaactaa actatgtgat ctcttggaat caattacaga 720 tttctccgtg ataggtatcg atgaaggaca gttctttcca gacattgttg aattagatcg 780 ataaaaatta attaattacc cgggtaccag gcctagatct gtcgacttcg agcttattta 840 tattccaaaa aaaaaaaata aaatttcaat ttttaagctt tcactaattc caaacccacc 900 cgctttttat agtaagtttt tcacccataa ataataaata caataattaa tttctcgtaa 960 aagtagaaaa tatattctaa tttattgcac ggtaaggaag tagatcataa ctcgagcatg 1020 ggagatcccg tcgttttaca acgtcgtgac tgggaaaacc ctggcgttac ccaacttaat 1080 cgccttgcag cacatccccc tttcgccagc tggcgtaata gcgaagaggc ccgcaccgat 1140 cgcccttccc aacagttgcg cagcctgaat ggcgaatggc gctttgcctg gtttccggca 1200 ccagaagcgg tgccggaaag ctggctggag tgcgatcttc ctgaggccga tactgtcgtc 1260 gtcccctcaa actggcagat gcacggttac gatgcgccca tctacaccaa cgtaacctat 1320 cccattacgg tcaatccgcc gtttgttccc acggagaatc cgacgggttg ttactcgctc 1380 acatttaatg ttgatgaaag ctggctacag gaaggccaga cgcgaattat ttttgatggc 1440 gttaactcgg cgtttcatct gtggtgcaac gggcgctggg tcggttacgg ccaggacagt 1500 cgtttgccgt ctgaatttga cctgagcgca tttttacgcg ccggagaaaa ccgcctcgcg 1560 gtgatggtgc tgcgttggag tgacggcagt tatctggaag atcaggatat gtggcggatg 1620 agcggcattt tccgtgacgt ctcgttgctg cataaaccga ctacacaaat cagcgatttc 1680 catgttgcca ctcgctttaa tgatgatttc agccgcgctg tactggaggc tgaagttcag 1740 atgtgcggcg agttgcgtga ctacctacgg gtaacagttt ctttatggca gggtgaaacg 1800 caggtcgcca gcggcaccgc gcctttcggc ggtgaaatta tcgatgagcg tggtggttat 1860 gccgatcgcg tcacactacg tctcaacgtc gaaaacccga aactgtggag cgccgaaatc 1920 ccgaatctct atcgtgcggt ggttgaactg cacaccgccg acggcacgct gattgaagca 1980 gaagcctgcg atgtcggttt ccgcgaggtg cggattgaaa atggtctgct gctgctgaac 2040 ggcaagccgt tgctgattcg aggcgttaac cgtcacgagc atcatcctct gcatggtcag 2100 gtcatggatg agcagacgat ggtgcaggat atcctgctga tgaagcagaa caactttaac 2160 gccgtgcgct gttcgcatta tccgaaccat ccgctgtggt acacgctgtg cgaccgctac 2220 ggcctgtatg tggtggatga agccaatatt gaaacccacg gcatggtgcc aatgaatcgt 2280 ctgaccgatg atccgcgctg gctaccggcg atgagcgaac gcgtaacgcg aatggtgcag 2340 cgcgatcgta atcacccgag tgtgatcatc tggtcgctgg ggaatgaatc aggccacggc 2400 gctaatcacg acgcgctgta tcgctggatc aaatctgtcg atccttcccg cccggtgcag 2460 tatgaaggcg gcggagccga caccacggcc accgatatta tttgcccgat gtacgcgcgc 2520 gtggatgaag accagccctt cccggctgtg ccgaaatggt ccatcaaaaa atggctttcg 2580 ctacctggag agacgcgccc gctgatcctt tgcgaatacg cccacgcgat gggtaacagt 2640 cttggcggtt tcgctaaata ctggcaggcg tttcgtcagt atccccgttt acagggcggc 2700 ttcgtctggg actgggtgga tcagtcgctg attaaatatg atgaaaacgg caacccgtgg 2760 tcggcttacg gcggtgattt tggcgatacg ccgaacgatc gccagttctg tatgaacggt 2820 ctggtctttg ccgaccgcac gccgcatcca gcgctgacgg aagcaaaaca ccagcagcag 2880 tttttccagt tccgtttatc cgggcaaacc atcgaagtga ccagcgaata cctgttccgt 2940 catagcgata acgagctcct gcactggatg gtggcgctgg atggtaagcc gctggcaagc 3000 ggtgaagtgc ctctggatgt cgctccacaa ggtaaacagt tgattgaact gcctgaacta 3060 ccgcagccgg agagcgccgg gcaactctgg ctcacagtac gcgtagtgca accgaacgcg 3120 accgcatggt cagaagccgg gcacatcagc gcctggcagc agtggcgtct ggcggaaaac 3180 ctcagtgtga cgctccccgc cgcgtcccac gccatcccgc atctgaccac cagcgaaatg 3240 gatttttgca tcgagctggg taataagcgt tggcaattta accgccagtc aggctttctt 3300 tcacagatgt ggattggcga taaaaaacaa ctgctgacgc cgctgcgcga tcagttcacc 3360 cgtgcaccgc tggataacga cattggcgta agtgaagcga cccgcattga ccctaacgcc 3420 tgggtcgaac gctggaaggc ggcgggccat taccaggccg aagcagcgtt gttgcagtgc 3480 acggcagata cacttgctga tgcggtgctg attacgaccg ctcacgcgtg gcagcatcag 3540 gggaaaacct tatttatcag ccggaaaacc taccggattg atggtagtgg tcaaatggcg 3600 attaccgttg atgttgaagt ggcgagcgat acaccgcatc cggcgcggat tggcctgaac 3660 tgccagctgg cgcaggtagc agagcgggta aactggctcg gattagggcc gcaagaaaac 3720 tatcccgacc gccttactgc cgcctgtttt gaccgctggg atctgccatt gtcagacatg 3780 tataccccgt acgtcttccc gagcgaaaac ggtctgcgct gcgggacgcg cgaattgaat 3840 tatggcccac accagtggcg cggcgacttc cagttcaaca tcagccgcta cagtcaacag 3900 caactgatgg aaaccagcca tcgccatctg ctgcacgcgg aagaaggcac atggctgaat 3960 atcgacggtt tccatatggg gattggtggc gacgactcct ggagcccgtc agtatcggcg 4020 gaattcagct gagcgccggt cgctaccatt accagttggt ctggtgtcaa aaataataat 4080 aaccgggcag gggggatcct tctgtgagcg tatggcaaac gaaggaaaaa tagttatagt 4140 agccgcactc gatgggacat ttcaacgtaa accgtttaat aatattttga atcttattcc 4200 attatctgaa atggtggtaa aactaactgc tgtgtgtatg aaatgcttta aggaggcttc 4260 cttttctaaa cgattgggtg aggaaaccga gatagaaata ataggaggta atgatatgta 4320 tcaatcggtg tgtagaaagt gttacatcga ctcataatat tatatttttt atctaaaaaa 4380 ctaaaaataa acattgatta aattttaata taatacttaa aaatggatgt tgtgtcgtta 4440 gataaaccgt ttatgtattt tgaggaaatt gataatgagt tagattacga accagaaagt 4500 gcaaatgagg tcgcaaaaaa actgccgtat caaggacagt taaaactatt actaggagaa 4560 ttattttttc ttagtaagtt acagcgacac ggtatattag atggtgccac cgtagtgtat 4620 ataggatctg ctcccggtac acatatacgt tatttgagag atcatttcta taatttagga 4680 gtgatcatca aatggatgct aattgacggc cgccatcatg atcctatttt aaatggattg 4740 cgtgatgtga ctctagtgac tcggttcgtt gatgaggaat atctacgatc catcaaaaaa 4800 caactgcatc cttctaagat tattttaatt tctgatgtga gatccaaacg aggaggaaat 4860 gaacctagta cggcggattt actaagtaat tacgctctac aaaatgtcat gattagtatt 4920 ttaaaccccg tggcgtctag tcttaaatgg agatgcccgt ttccagatca atggatcaag 4980 gacttttata tcccacacgg taataaaatg ttacaacctt ttgctccttc atattcagct 5040 gaaatgagat tattaagtat ttataccggt gagaacatga gactgactcg ggccgcgttg 5100 ctggcgtttt tccataggct ccgcccccct gacgagcatc acaaaaatcg acgctcaagt 5160 cagaggtggc gaaacccgac aggactataa agataccagg cgtttccccc tggaagctcc 5220 ctcgtgcgct ctcctgttcc gaccctgccg cttaccggat acctgtccgc ctttctccct 5280 tcgggaagcg tggcgctttc tcaatgctca cgctgtaggt atctcagttc ggtgtaggtc 5340 gttcgctcca agctgggctg tgtgcacgaa ccccccgttc agcccgaccg ctgcgcctta 5400 tccggtaact atcgtcttga gtccaacccg gtaagacacg acttatcgcc actggcagca 5460 gccactggta acaggattag cagagcgagg tatgtaggcg gtgctacaga gttcttgaag 5520 tggtggccta actacggcta cactagaagg acagtatttg gtatctgcgc tctgctgaag 5580 ccagttacct tcggaaaaag agttggtagc tcttgatccg gcaaacaaac caccgctggt 5640 agcggtggtt tttttgtttg caagcagcag attacgcgca gaaaaaaagg atctcaagaa 5700 gatcctttga tcttttctac ggggtctgac gctcagtgga acgaaaactc acgttaaggg 5760 attttggtca tgagattatc aaaaaggatc ttcacctaga tccttttaaa ttaaaaatga 5820 agttttaaat caatctaaag tatatatgag taaacttggt ctgacagtta ccaatgctta 5880 atcagtgagg cacctatctc agcgatctgt ctatttcgtt catccatagt tgcctgactc 5940 cccgtcgtgt agataactac gatacgggag ggcttaccat ctggccccag tgctgcaatg 6000 ataccgcgag acccacgctc accggctcca gatttatcag caataaacca gccagccgga 6060 agggccgagc gcagaagtgg tcctgcaact ttatccgcct ccatccagtc tattaattgt 6120 tgccgggaag ctagagtaag tagttcgcca gttaatagtt tgcgcaacgt tgttgccatt 6180 gctgcaggca tcgtggtgtc acgctcgtcg tttggtatgg cttcattcag ctccggttcc 6240 caacgatcaa ggcgagttac atgatccccc atgttgtgca aaaaagcggt tagctccttc 6300 ggtcctccga tcgttgtcag aagtaagttg gccgcagtgt tatcactcat ggttatggca 6360 gcactgcata attctcttac tgtcatgcca tccgtaagat gcttttctgt gactggtgag 6420 tactcaacca agtcattctg agaatagtgt atgcggcgac cgagttgctc ttgcccggcg 6480 tcaacacggg ataataccgc gccacatagc agaactttaa aagtgctcat cattggaaaa 6540 cgttcttcgg ggcgaaaact ctcaaggatc ttaccgctgt tgagatccag ttcgatgtaa 6600 cccactcgtg cacccaactg atcttcagca tcttttactt tcaccagcgt ttctgggtga 6660 gcaaaaacag gaaggcaaaa tgccgcaaaa aagggaataa gggcgacacg gaaatgttga 6720 atactcatac tcttcctttt tcaatattat tgaagcattt atcagggtta ttgtctcatg 6780 agcggataca tatttgaatg tatttagaaa aataaacaaa taggggttcc gcgcacattt 6840 ccccgaaaag tgccacctga cgtctaagaa accattatta tcatgacatt aacctataaa 6900 aataggcgta tcacgaggcc ctttcgtctt cgaataaata cctgtgacgg aagatcactt 6960 cgcagaataa ataaatcctg gtgtccctgt tgataccggg aagccctggg ccaacttttg 7020 gcgaaaatga gacgttgatc ggcacgtaag aggttccaac tttcaccata atgaaataag 7080 atcactaccg ggcgtatttt ttgagttatc gagattttca ggagctaagg aagctaaaat 7140 ggagaaaaaa atcactggat ataccaccgt tgatatatcc caatggcatc gtaaagaaca 7200 ttttgaggca tttcagtcag ttgctcaatg tacctataac cagaccgttc ag 7252 28 7387 DNA Artificial Sequence, primer misc_feature (1)..(7387) plasmid vector 28 cctcctgaaa aactggaatt taatacacca tttgtgttca tcatcagaca tgatattact 60 ggatttatat tgtttatggg taaggtagaa tctccttaat atgggtacgg tgtaaggaat 120 cattatttta tttatattga tgggtacgtg aaatctgaat tttcttaata aatattattt 180 ttattaaatg tgtatatgtt gttttgcgat agccatgtat ctactaatca gatctattag 240 agatattatt aattctggtg caatatgaca aaaattatac actaattagc gtctcgtttc 300 agacatggat ctgtcacgaa ttaatacttg gaagtctaag cagctgaaaa gctttctctc 360 tagcaaagat gcatttaagg cggatgtcca tggacatagt gccttgtatt atgcaatagc 420 tgataataac gtgcgtctag tatgtacgtt gttgaacgct ggagcattga aaaatcttct 480 agagaatgaa tttccattac atcaggcagc cacattggaa gataccaaaa tagtaaagat 540 tttggctatt cagtggactg gatgattcga ggtacccgat cccccctgcc cggttattat 600 tatttttgac accagaccaa ctggtaatgg tagcgaccgg cgctcagctg aattccgccg 660 atactgacgg gctccaggag tcgtcgccac caatccccat atggaaaccg tcgatattca 720 gccatgtgcc ttcttccgcg tgcagcagat ggcgatggct ggtttccatc agttgctgtt 780 gactgtagcg gctgatgttg aactggaagt cgccgcgcca ctggtgtggg ccataattca 840 attcgcgcgt cccgcagcgc agaccgtttt cgctcgggaa gacgtacggg gtatacatgt 900 ctgacaatgg cagatcccag cggtcaaaac aggcggcagt aaggcggtcg ggatagtttt 960 cttgcggccc taatccgagc cagtttaccc gctctgctac ctgcgccagc tggcagttca 1020 ggccaatccg cgccggatgc ggtgtatcgc tcgccacttc aacatcaacg gtaatcgcca 1080 tttgaccact accatcaatc cggtaggttt tccggctgat aaataaggtt ttcccctgat 1140 gctgccacgc gtgagcggtc gtaatcagca ccgcatcagc aagtgtatct gccgtgcact 1200 gcaacaacgc tgcttcggcc tggtaatggc ccgccgcctt ccagcgttcg acccaggcgt 1260 tagggtcaat gcgggtcgct tcacttacgc caatgtcgtt atccagcggt gcacgggtga 1320 actgatcgcg cagcggcgtc agcagttgtt ttttatcgcc aatccacatc tgtgaaagaa 1380 agcctgactg gcggttaaat tgccaacgct tattacccag ctcgatgcaa aaatccattt 1440 cgctggtggt cagatgcggg atggcgtggg acgcggcggg gagcgtcaca ctgaggtttt 1500 ccgccagacg ccactgctgc caggcgctga tgtgcccggc ttctgaccat gcggtcgcgt 1560 tcggttgcac tacgcgtact gtgagccaga gttgcccggc gctctccggc tgcggtagtt 1620 caggcagttc aatcaactgt ttaccttgtg gagcgacatc cagaggcact tcaccgcttg 1680 ccagcggctt accatccagc gccaccatcc agtgcaggag ctcgttatcg ctatgacgga 1740 acaggtattc gctggtcact tcgatggttt gcccggataa acggaactgg aaaaactgct 1800 gctggtgttt tgcttccgtc agcgctggat gcggcgtgcg gtcggcaaag accagaccgt 1860 tcatacagaa ctggcgatcg ttcggcgtat cgccaaaatc accgccgtaa gccgaccacg 1920 ggttgccgtt ttcatcatat ttaatcagcg actgatccac ccagtcccag acgaagccgc 1980 cctgtaaacg gggatactga cgaaacgcct gccagtattt agcgaaaccg ccaagactgt 2040 tacccatcgc gtgggcgtat tcgcaaagga tcagcgggcg cgtctctcca ggtagcgaaa 2100 gccatttttt gatggaccat ttcggcacag ccgggaaggg ctggtcttca tccacgcgcg 2160 cgtacatcgg gcaaataata tcggtggccg tggtgtcggc tccgccgcct tcatactgca 2220 ccgggcggga aggatcgaca gatttgatcc agcgatacag cgcgtcgtga ttagcgccgt 2280 ggcctgattc attccccagc gaccagatga tcacactcgg gtgattacga tcgcgctgca 2340 ccattcgcgt tacgcgttcg ctcatcgccg gtagccagcg cggatcatcg gtcagacgat 2400 tcattggcac catgccgtgg gtttcaatat tggcttcatc caccacatac aggccgtagc 2460 ggtcgcacag cgtgtaccac agcggatggt tcggataatg cgaacagcgc acggcgttaa 2520 agttgttctg cttcatcagc aggatatcct gcaccatcgt ctgctcatcc atgacctgac 2580 catgcagagg atgatgctcg tgacggttaa cgcctcgaat cagcaacggc ttgccgttca 2640 gcagcagcag accattttca atccgcacct cgcggaaacc gacatcgcag gcttctgctt 2700 caatcagcgt gccgtcggcg gtgtgcagtt caaccaccgc acgatagaga ttcgggattt 2760 cggcgctcca cagtttcggg ttttcgacgt tgagacgtag tgtgacgcga tcggcataac 2820 caccacgctc atcgataatt tcaccgccga aaggcgcggt gccgctggcg acctgcgttt 2880 caccctgcca taaagaaact gttacccgta ggtagtcacg caactcgccg cacatctgaa 2940 cttcagcctc cagtacagcg cggctgaaat catcattaaa gcgagtggca acatggaaat 3000 cgctgatttg tgtagtcggt ttatgcagca acgagacgtc acggaaaatg ccgctcatcc 3060 gccacatatc ctgatcttcc agataactgc cgtcactcca acgcagcacc atcaccgcga 3120 ggcggttttc tccggcgcgt aaaaatgcgc tcaggtcaaa ttcagacggc aaacgactgt 3180 cctggccgta accgacccag cgcccgttgc accacagatg aaacgccgag ttaacgccat 3240 caaaaataat tcgcgtctgg ccttcctgta gccagctttc atcaacatta aatgtgagcg 3300 agtaacaacc cgtcggattc tccgtgggaa caaacggcgg attgaccgta atgggatagg 3360 ttacgttggt gtagatgggc gcatcgtaac cgtgcatctg ccagtttgag gggacgacga 3420 cagtatcggc ctcaggaaga tcgcactcca gccagctttc cggcaccgct tctggtgccg 3480 gaaaccaggc aaagcgccat tcgccattca ggctgcgcaa ctgttgggaa gggcgatcgg 3540 tgcgggcctc ttcgctatta cgccagctgg cgaaaggggg atgtgctgca aggcgattaa 3600 gttgggtaac gccagggttt tcccagtcac gacgttgtaa aacgacggga tctcccatgc 3660 tcgagttatg atctacttcc ttaccgtgca ataaattaga atatattttc tacttttacg 3720 agaaattaat tattgtattt attatttatg ggtgaaaaac ttactataaa aagcgggtgg 3780 gtttggaatt agtgaaagct gggagatctg gcgcgcctgc agagaattcg tttaaacgga 3840 tcccgagctt atttatattc caaaaaaaaa aaataaaatt tcaattttta agctggggat 3900 cctctagagt cgacctgcag gcatgctcga gcggccgcca gtgtgatgga tatctgcaga 3960 attcggcttg gggggctgca ggtggatgcg atcatgacgt cctctgcaat ggataacaat 4020 gaacctaaag tactagaaat ggtatatgat gctacaattt tacccgaagg tagtagcatg 4080 gattgtataa acagacacat caatatgtgt atacaacgca cctatagttc tagtataatt 4140 gccatattgg atagattcct aatgatgaac aaggatgaac taaataatac acagtgtcat 4200 ataattaaag aatttatgac atacgaacaa atggcgattg accattatgg agaatatgta 4260 aacgctattc tatatcaaat tcgtaaaaga cctaatcaac atcacaccat taatctgttt 4320 aaaaaaataa aaagaacccg gtatgacact tttaaagtgg atcccgtaga attcgtaaaa 4380 aaagttatcg gatttgtatc tatcttgaac aaatataaac cggtttatag ttacgtcctg 4440 tacgagaacg tcctgtacga tgagttcaaa tgtttcattg actacgtgga aactaagtat 4500 ttctaaaatt aatgatgcat taatttttgt attgattctc aatcctaaaa actaaaatat 4560 gaataagtat taaacatagc ggtgtactaa ttgatttaac ataaaaaata gttgttaact 4620 aatcatgagg actctactta ttagatatat tctttggaga aatgacaacg atcaaaccgg 4680 gcatgcaagc ttgtctccct atagtgagtc gtattagagc ttggcgtaat catggtcata 4740 gctgtttcct gtgtgaaatt gttatccgct cacaattcca cacaacatac gagccggaag 4800 cataaagtgt aaagcctggg gtgcctaatg agtgagctaa ctcacattaa ttgcgttgcg 4860 ctcactgccc gctttcgagt cgggaaacct gtcgtgccag ctgcattaat gaatcggcca 4920 acgcgcgggg agaggcggtt tgcgtattgg gcgctcttcc gcttcctcgc tcactgactc 4980 gctgcgctcg gtcgttcggc tgcggcgagc ggtatcagct cactcaaagg cggtaatacg 5040 gttatccaca gaatcagggg ataacgcagg aaagaacatg tgagcaaaag gccagcaaaa 5100 ggccaggaac cgtaaaaagg ccgcgttgct ggcgtttttc gataggctcc gcccccctga 5160 cgagcatcac aaaaatcgac gctcaagtca gaggtggcga aacccgacag gactataaag 5220 ataccaggcg tttccccctg gaagctccct cgtgcgctct cctgttccga ccctgccgct 5280 taccggatac ctgtccgcct ttctcccttc gggaagcgtg gcgctttctc atagctcacg 5340 ctgtaggtat ctcagttcgg tgtaggtcgt tcgctccaag ctgggctgtg tgcacgaacc 5400 ccccgttcag cccgaccgct gcgccttatc cggtaactat cgtcttgagt ccaacccggt 5460 aagacacgac ttatcgccac tggcagcagc cactggtaac aggattagca gagcgaggta 5520 tgtaggcggt gctacagagt tcttgaagtg gtggcctaac tacggctaca ctagaaggac 5580 agtatttggt atctgcgctc tgctgaagcc agttaccttc ggaaaaagag ttggtagctc 5640 ttgatccggc aaacaaacca ccgctggtag cggtggtttt tttgtttgca agcagcagat 5700 tacgcgcaga aaaaaaggat ctcaagaaga tcctttgatc ttttctacgg ggtctgacgc 5760 tcagtggaac gaaaactcac gttaagggat tttggtcatg agattatcaa aaaggatctt 5820 cacctagatc cttttaaatt aaaaatgaag ttttaaatca atctaaagta tatatgagta 5880 aacttggtct gacagttacc aatgcttaat cagtgaggca cctatctcag cgatctgtct 5940 atttcgttca tccatagttg cctgactccc cgtcgtgtag ataactacga tacgggaggg 6000 cttaccatct ggccccagtg ctgcaatgat accgcgagac ccacgctcac cggctccaga 6060 tttatcagca ataaaccagc cagccggaag ggccgagcgc agaagtggtc ctgcaacttt 6120 atccgcctcc atccagtcta ttaattgttg ccgggaagct agagtaagta gttcgccagt 6180 taatagtttg cgcaacgttg ttggcattgc tacaggcatc gtggtgtcac gctcgtcgtt 6240 tggtatggct tcattcagct ccggttccca acgatcaagg cgagttacat gatcccccat 6300 gttgtgcaaa aaagcggtta gctccttcgg tcctccgatc gttgtcagaa gtaagttggc 6360 cgcagtgtta tcactcatgg ttatggcagc actgcataat tctcttactg tcatgccatc 6420 cgtaagatgc ttttctgtga ctggtgagta ctcaaccaag tcattctgag aatagtgtat 6480 gcggcgaccg agttgctctt gcccggcgtc aatacgggat aataccgcgc cacatagcag 6540 aactttaaaa gtgctcatca ttggaaaacg ttcttcgggg cgaaaactct caaggatctt 6600 accgctgttg agatccagtt cgatgtaacc cactcgtgca cccaactgat cttcagcatc 6660 ttttactttc accagcgttt ctgggtgagc aaaaacagga aggcaaaatg ccgcaaaaaa 6720 gggaataagg gcgacacgga aatgttgaat actcatactc ttcctttttc aatattattg 6780 aagcatttat cagggttatt gtctcatgag cggatacata tttgaatgta tttagaaaaa 6840 taaacaaata ggggttccgc gcacatttcc ccgaaaagtg ccacctgacg tctaagaaac 6900 cattattatc atgacattaa cctataaaaa taggcgtatc acgaggccct ttcgtctcgc 6960 gcgtttcggt gatgacggtg aaaacctctg acacatgcag ctcccggaga cggtcacagc 7020 ttgtctgtaa gcggatgccg ggagcagaca agcccgtcag ggcgcgtcag cgggtgttgg 7080 cgggtgtcgg ggctggctta actatgcggc atcagagcag attgtactga gagtgcacca 7140 tatgcggtgt gaaataccgc acagatgcgt aaggagaaaa taccgcatca ggcgccattc 7200 gccattcagg ctgcgcaact gttgggaagg gcgatcggtg cgggcctctt cgctattacg 7260 ccagctggcg aaagggggat gtgctgcaag gcgattaagt tgggtaacgc cagggttttc 7320 ccagtcacga cgttgtaaaa cgacggccag tgaattggat ttaggtgaca ctatagaata 7380 cgaattc 7387 29 27 DNA Artificial Sequence, primer misc_feature (1)..(27) primer 29 gcatggacct gtggggtttt tatgagg 27 30 29 DNA Artificial Sequence, primer misc_feature (1)..(29) primer 30 gcatgagctc tgtaggatct cgaacagac 29 31 33 DNA Artificial Sequence, primer misc_feature (1)..(33) primer 31 gactacgact agtgtatgtt tagaaaaaca agg 33 32 32 DNA Artificial Sequence misc_feature (1)..(32) primer 32 ctaggctact agtactgtag gatctcgaac ag 32 33 33 DNA Artificial Sequence, primer misc_feature (1)..(33) primer 33 gggctatatg agatcttgaa taataaaatg tgt 33 34 14 DNA Artificial Sequence, primer misc_feature (1)..(14) primer 34 tattaataac tagt 14 35 42 DNA Artificial Sequence, primer misc_feature (1)..(42) primer 35 gctacgcaga gctcgtttag tgaaccgggc actcagattc tg 42 36 36 DNA Artificial Sequence, primer misc_feature (1)..(36) primer 36 gctgagctct agagtccttt tcttttacaa agttgg 36 37 31 DNA Artificial Sequence, primer misc_feature (1)..(31) primer 37 gtcgctgagg tcgacaaggc aaagagaaga g 31 38 31 DNA Artificial Sequence, primer misc_feature (1)..(31) primer 38 gaccggtacc gtcgacaagg cacagcagtg g 31 39 32 DNA Artificial Sequence, primer misc_feature (1)..(32) primer 39 ttctgtcgac gaatcccagg gggaatctca ac 32 40 35 DNA Artificial Sequence, primer misc_feature (1)..(35) primer 40 gtcaccttcc agagggccct ggctaagcat aacag 35 41 35 DNA Artificial Sequence, primer misc_feature (1)..(35) primer 41 ctgttatgct tagccagggc cctctggaag gtgac 35 42 28 DNA Artificial Sequence, primer misc_feature (1)..(28) primer 42 aattgctgac ccccaaaata gccataag 28 43 36 DNA Artificial Sequence, primer misc_feature (1)..(36) primer 43 ccatgcacgt ctgcagccag catggcagaa tcgaag 36 44 30 DNA Artificial Sequence, primer misc_feature (1)..(30) primer 44 cctgaggatc tattttccac cagtcatttc 30 45 29 DNA Artificial Sequence, primer misc_feature (1)..(29) primer 45 gtggaaaata gatcctcagg gccctctgg 29 46 34 DNA Artificial Sequence, primer misc_feature (1)..(34) primer 46 gcagtgccgg atcctcataa atgtttcctc cttc 34 47 24 DNA Artificial Sequence, primer misc_feature (1)..(24) primer 47 gacaccatgg gaagtattta tcac 24 48 35 DNA Artificial Sequence, primer misc_feature (1)..(35) primer 48 cctgggattc atatcaaacc ttataacaaa tattg 35 49 22 DNA Artificial Sequence, primer misc_feature (1)..(22) primer 49 tcctgctaag cataacagaa ac 22 50 29 DNA Artificial Sequence, primer misc_feature (1)..(29) primer 50 ggtttgatat gaatcccagg gggaatctc 29 51 22 DNA Artificial Sequence, primer misc_feature (1)..(22) primer 51 accccgtacg tcttcccgag cg 22 52 39 DNA Artificial Sequence, primer misc_feature (1)..(39) primer 52 gttattaatt aatggaggaa taattgaaga aggatatac 39 53 31 DNA Artificial Sequence, primer misc_feature (1)..(31) primer 53 tcttctgcag gtcctgatcc ttgcttagtg c 31 54 41 DNA Artificial Sequence, primer misc_feature (1)..(41) primer 54 gaccatgtta cccctttacc attaactccc taatatcaaa c 41 55 44 DNA Artificial Sequence, primer misc_feature (1)..(44) primer 55 gtaaaggggt aacatggtca gcatcgcatt ctacggggga atcc 44 56 38 DNA Artificial Sequence, primer misc_feature (1)..(38) primer 56 ccatgcacgt ctcgagccag catgggagac cctttgac 38 57 37 DNA Artificial Sequence, primer misc_feature (1)..(37) primer 57 cgagctagag gtcgactcaa tttggtttat tagtaac 37 58 32 DNA Artificial Sequence, primer misc_feature (1)..(32) primer 58 gcaatggaat gacatccctc agctgccagt cc 32 59 42 DNA Artificial Sequence, primer misc_feature (1)..(42) primer 59 gggatgtcat tccattgcca ccatgggaag tatttatcac ta 42 60 34 DNA Artificial Sequence, primer misc_feature (1)..(34) primer 60 gtcgagcacg cgtttgccta gcaacatgag ctag 34 61 34 DNA Artificial Sequence, primer misc_feature (1)..(34) primer 61 gtcgagccaa ttgttgccta gcaacatgag ctag 34 62 28 DNA Artificial Sequence misc_feature (1)..(28) P7.5E sequence 62 aaaagtagaa aatatattct aatttatt 28 63 19 DNA Artificial Sequence promoter (1)..(19) T7 promter 63 taatacgact cactatagg 19 64 48 DNA Artificial Sequence terminator (1)..(48) T7 terminator 64 ctagcataac cccttggggc ctctaaacgg gtcttgaggg gttttttg 48

Claims (29)

1. A retroviral vector derived from a non-primate lentivirus genome comprising a deleted gag gene wherein the deletion in gag removes one or more nucleotides downstream of nucleotide 350 of the gag coding sequence.
2. A retroviral vector according to claim 1 wherein the deletion extends from nucleotide 350 to at least the C-terminus of the gag-pol coding region.
3. A retroviral vector according to claim 1 or claim 2 wherein the deletion additionally removes nucleotide 300 of the gag coding region.
4. A retroviral vector according to claim 1 wherein the deletion retains the first 150 nucleotides of the gag coding region.
5. A retroviral vector according to claim 1 wherein the deletion retains the first 109 nucleotides of the gag coding region.
6. A retroviral vector according to claim 1 wherein the deletion retains only the first 2 nucleotides of the gag coding region.
7. A retroviral vector derived from a non-primate lentivirus genome wherein one or more accessory genes are absent from the non-primate lentivirus genome.
8. A retroviral vector according to claim 7 wherein the accessory genes are selected from dUTPase, S2, env and tat.
9. A retroviral vector derived from a lentivirus genome wherein the non-primate lentivirus genome lacks the tat gene but includes the leader sequences between the end of the 5′ LTR and the ATG of gag.
10. A retroviral vector according to any preceding claim which comprises at least one component from an equinine lentivirus.
11. A retroviral vector according to claim 10 wherein the equinine lentivirus is EIAV.
12. A retroviral vector according to claim 11 wherein the retroviral vector is substantially derived from EIAV.
13. A retroviral vector production system for producing the retroviral vector of any preceding claim comprising a packaging cell comprising a gag-pol gene from a non-primate lentivirus and an envelope gene.
14. A retroviral vector produced by the production system of claim 13.
15. A hybrid viral vector system comprising a primary viral vector derived from a poxvirus and a second viral vector derived from a retrovirus.
16. A retroviral particle obtainable from the retroviral vector of any one of claims 1 to 12 or claim 14 or 15.
17. A cell transfected or transduced with a retroviral vector according to any one of claims 1 to 12 or claim 14 or 15 or a retroviral particle of claim 16.
18. A retroviral vector according to any one of claims 1 to 12 or claim 14 or 15 or a retroviral particle of claim 16 or a cell according to claim 17 for use in medicine.
19. Use of a retroviral vector according to any one of claims 1 to 12 or claim 14 or 15 or a retroviral particle of claim 16 or a cell according to claim 17 for the manufacture of a pharmaceutical composition to deliver an NOI to a target site in need of same.
20. A method comprising transfecting or transducing a cell with a retroviral vector according to any one of claims 1 to 12 or claim 14 or 15 or a retroviral particle of claim 16 or by use of a cell according to claim 17.
21. A delivery system in the form of a retroviral vector according to any one of claims 1 to 12 or claim 14 or 15 or a retroviral particle of claim 16 or a cell according to claim 17.
22. A delivery system for a retroviral vector according to any one of claims 1 to 12 or claim 14 or 15 or a retroviral particle of claim 16 or a cell according to claim 17 wherein the delivery system comprises a non-retroviral expression vector, an adenovirus and/or a plasmid.
23. A retroviral vector substantially as hereinbefore described with reference to the accompanying Figures.
24. A retroviral vector production system substantially as hereinbefore described with reference to the accompanying Figures.
25. A retroviral particle substantially as hereinbefore described with reference to the accompanying Figures.
26. A cell transfected or transduced with a retroviral vector substantially as hereinbefore described with reference to the accompanying Figures.
27. Use of a retroviral vector substantially as hereinbefore described with reference to the accompanying Figures.
28. A method comprising transfecting or transducing a cell with a retroviral vector substantially as hereinbefore described with reference to the accompanying Figures.
29. A delivery system in the form of a retroviral vector substantially as hereinbefore described with reference to the accompanying Figures.
US09/867,947 1996-10-17 2001-05-29 Retroviral vectors Abandoned US20030147907A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US09/867,947 US20030147907A1 (en) 1997-12-22 2001-05-29 Retroviral vectors
US11/154,421 US20060024274A1 (en) 1997-12-22 2005-06-15 Retroviral vectors
US11/583,427 US20070213290A1 (en) 1996-10-17 2006-10-19 Neurite regeneration

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GBGB9727135.7A GB9727135D0 (en) 1997-12-22 1997-12-22 Non-primate lentiviral vectors
GB9811037.2 1998-05-22
GBGB9811037.2A GB9811037D0 (en) 1998-05-22 1998-05-22 Proretoviral vectors
GB9727135.7 1998-05-22
PCT/GB1998/003876 WO1999032646A1 (en) 1997-12-22 1998-12-22 Equine infectious anaemia virus (eiav) based
US09/238,356 US6312683B1 (en) 1997-12-22 1999-01-27 Equine infectious anemia virus vectors
US09/867,947 US20030147907A1 (en) 1997-12-22 2001-05-29 Retroviral vectors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/238,356 Division US6312683B1 (en) 1996-10-17 1999-01-27 Equine infectious anemia virus vectors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/154,421 Continuation US20060024274A1 (en) 1996-10-17 2005-06-15 Retroviral vectors

Publications (1)

Publication Number Publication Date
US20030147907A1 true US20030147907A1 (en) 2003-08-07

Family

ID=26312829

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/238,356 Expired - Lifetime US6312683B1 (en) 1996-10-17 1999-01-27 Equine infectious anemia virus vectors
US09/867,947 Abandoned US20030147907A1 (en) 1996-10-17 2001-05-29 Retroviral vectors
US11/154,421 Abandoned US20060024274A1 (en) 1996-10-17 2005-06-15 Retroviral vectors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/238,356 Expired - Lifetime US6312683B1 (en) 1996-10-17 1999-01-27 Equine infectious anemia virus vectors

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/154,421 Abandoned US20060024274A1 (en) 1996-10-17 2005-06-15 Retroviral vectors

Country Status (12)

Country Link
US (3) US6312683B1 (en)
EP (2) EP1042493A1 (en)
JP (1) JP4578678B2 (en)
CN (1) CN1322137C (en)
AT (1) ATE528405T1 (en)
AU (1) AU760287B2 (en)
CA (1) CA2315269A1 (en)
CY (1) CY1112196T1 (en)
DK (1) DK1895010T3 (en)
ES (1) ES2373406T3 (en)
PT (1) PT1895010E (en)
WO (1) WO1999032646A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100003746A1 (en) * 2007-02-12 2010-01-07 Lesch Hanna P Production of Lentiviral Vectors
WO2022204351A3 (en) * 2021-03-24 2022-11-24 National Resilience, Inc. Systems and methods for cell-free polypeptide synthesis

Families Citing this family (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040266715A1 (en) * 1999-03-31 2004-12-30 Wong Liang Fong Neurite regeneration
US20070213290A1 (en) * 1996-10-17 2007-09-13 Kingsman Alan J Neurite regeneration
DK1895010T3 (en) * 1997-12-22 2011-11-21 Oxford Biomedica Ltd Vectors based on viruses for infectious horse anemia (eiav)
GB9803351D0 (en) 1998-02-17 1998-04-15 Oxford Biomedica Ltd Anti-viral vectors
AU2001240078A1 (en) * 2000-03-07 2001-09-17 St. Jude Children's Research Hospital Highly efficient gene transfer into human repopulating stem cells by rd114 pseudotyped retroviral vector particles
US6849454B2 (en) 2000-03-07 2005-02-01 St. Jude Children's Research Hospital Highly efficient gene transfer into human repopulating stem cells by RD114 pseudotyped retroviral vector particles
EP1268835B1 (en) * 2000-03-30 2008-01-16 Oxford Biomedica (UK) Limited Retinoic acid receptor beta-2 and gene therapy vectors for the treatment of neurological disorders
GB0009760D0 (en) * 2000-04-19 2000-06-07 Oxford Biomedica Ltd Method
US6585978B1 (en) * 2000-09-09 2003-07-01 Akzo Nobel EIA vaccine and diagnostic
BR0113745A (en) * 2000-09-09 2004-03-02 Akzo Nobel Nv Vaccine, diagnostic test and methods for immunizing mammals and preparing a vaccine
US20040018211A1 (en) * 2002-06-26 2004-01-29 Ronald Montelaro EIA vaccine and diagnostic
US6461616B1 (en) * 2000-09-09 2002-10-08 Akzo Nobel Nv EIAV p26 deletion vaccine and diagnostic
AU2001291849B2 (en) * 2000-09-14 2005-11-24 Ares Trading S.A. Use of IL-6R/IL-6 chimera in huntington's disease
GB0024550D0 (en) 2000-10-06 2000-11-22 Oxford Biomedica Ltd
US20030175972A1 (en) * 2000-10-20 2003-09-18 Daley George Q. Expression vectors and uses thereof
US7575924B2 (en) 2000-11-13 2009-08-18 Research Development Foundation Methods and compositions relating to improved lentiviral vectors and their applications
US6712612B1 (en) 2000-12-12 2004-03-30 Genecure Llc Safe and stable retroviral helper cell line and related compositions and methods
CA2344208A1 (en) 2001-04-30 2002-10-30 Oxford Biomedica (Uk) Limited Method
CA2456169C (en) * 2001-08-02 2012-05-22 Didier Trono Methods and compositions relating to improved lentiviral vector production systems
EP1438399A4 (en) * 2001-09-28 2005-09-14 Univ North Carolina Paramyxoviruses as gene transfer vectors to lung cells
KR20040054699A (en) * 2001-10-02 2004-06-25 엥스띠뛰 끌레이톤 드 라 러쉐르쉬 Methods and compositions relating to restricted expression lentiviral vectors and their applications
US20030121062A1 (en) * 2001-12-21 2003-06-26 Oxford Biomedica (Uk) Limited Transgenic organism
JP2005512598A (en) * 2001-12-21 2005-05-12 オックスフォード バイオメディカ (ユーケー) リミテッド Method for producing transgenic organism using lentiviral expression vector such as EIAV
US9738907B2 (en) 2002-02-01 2017-08-22 Oxford Biomedica (Uk) Limited Viral vector
DK1504108T3 (en) 2002-02-01 2013-06-10 Oxford Biomedica Ltd lentiviral vector
GB0215287D0 (en) * 2002-07-02 2002-08-14 Oxford Biomedica Ltd 5T4 antigen expression
US7662625B2 (en) * 2002-07-02 2010-02-16 Cancer Research Technology Limited Methods for detecting the differentiation status of cells using 5T4 antigen expression
EP1534847B1 (en) * 2002-09-03 2015-10-28 Oxford Biomedica (UK) Limited Retroviral vector and stable packaging cell lines
GB0220467D0 (en) 2002-09-03 2002-10-09 Oxford Biomedica Ltd Composition
US20050251872A1 (en) * 2002-09-06 2005-11-10 Bear James E Lentiviral vectors, related reagents, and methods of use thereof
DK1563069T3 (en) * 2002-11-22 2012-07-23 Inst Clayton De La Rech COMPOSITIONS AND SYSTEMS FOR REGULATION
EP2390352A1 (en) 2003-03-18 2011-11-30 Quantum Genetics Ireland Limited Systems and methods for improving protein and milk production of dairy herds
US7468273B2 (en) 2003-05-01 2008-12-23 Meial Limited Canine GHRH gene, polypeptides and methods of use
AU2004289953B2 (en) 2003-06-18 2008-09-25 Genelux Corporation Modified recombinant vaccina viruses and other microorganisms, uses thereof
WO2005049794A2 (en) 2003-11-13 2005-06-02 University Of Georgia Research Foundation, Inc. Methods of characterizing infectious bursal disease virus
CA2556911C (en) 2004-02-19 2013-07-30 The Governors Of The University Of Alberta Leptin promoter polymorphisms and uses thereof
FR2870126B1 (en) 2004-05-17 2009-07-17 Pasteur Institut RECOMBINANT LENTIVIRAL VECTOR FOR EXPRESSION OF FLAVIVIRIDAE PROTEIN AND ITS APPLICATIONS AS VACCINE
US20060233757A1 (en) * 2004-08-27 2006-10-19 Wendy Maury Vectors with viral insulators
EP1881845B1 (en) 2005-04-25 2010-03-24 Merial Ltd. Nipah virus vaccines
US20080241184A1 (en) 2005-08-25 2008-10-02 Jules Maarten Minke Canine influenza vaccines
US7771995B2 (en) 2005-11-14 2010-08-10 Merial Limited Plasmid encoding human BMP-7
CA2629522A1 (en) 2005-11-14 2007-05-18 Merial Limited Gene therapy for renal failure
US7862821B2 (en) 2006-06-01 2011-01-04 Merial Limited Recombinant vaccine against bluetongue virus
EP2415783B1 (en) 2006-10-16 2016-12-14 Genelux Corporation Modified vaccinia virus strains for use in a diagnostic and therapeutic method
CN101705246B (en) * 2007-04-29 2012-06-27 中国农业科学院哈尔滨兽医研究所 Lentiviral gene transfer vector, preparation method and application thereof
WO2009065080A1 (en) 2007-11-16 2009-05-22 San Diego State University Research Foundation Compositions and method for manipulating pim-1 activity in circulatory system cells
CA2757030C (en) 2009-04-03 2019-01-15 Merial Limited Vectors comprising newcastle disease viruses and compositions thereof
CA2809127C (en) 2010-08-31 2019-04-02 Merial Limited Newcastle disease virus vectored herpesvirus vaccines
WO2012090073A2 (en) 2010-12-30 2012-07-05 The Netherlands Cancer Institute Methods and compositions for predicting chemotherapy sensitivity
WO2012138789A2 (en) 2011-04-04 2012-10-11 Netherlands Cancer Institute Methods and compositions for predicting resistance to anticancer treatment
WO2012138783A2 (en) 2011-04-04 2012-10-11 Netherlands Cancer Institute Methods and compositions for predicting resistance to anticancer treatment
AU2012245395A1 (en) 2011-04-20 2013-11-14 Merial, Inc. Adjuvanted rabies vaccine with improved viscosity profile
CA2834288A1 (en) 2011-04-25 2012-11-01 Advanced Bioscience Laboratories, Inc. Truncated hiv envelope proteins (env), methods and compositions related thereto
WO2012166493A1 (en) 2011-06-01 2012-12-06 Merial Limited Needle-free administration of prrsv vaccines
WO2013093629A2 (en) 2011-12-20 2013-06-27 Netherlands Cancer Institute Modular vaccines, methods and compositions related thereto
GB201202516D0 (en) 2012-02-13 2012-03-28 Ucl Business Plc Materials and methods relating to packaging cell lines
WO2013138776A1 (en) 2012-03-16 2013-09-19 Merial Limited Novel methods for providing long-term protective immunity against rabies in animals, based upon administration of replication-deficient flavivirus expressing rabies g
WO2014093965A1 (en) * 2012-12-14 2014-06-19 Case Western Reserve University Genomic rna packaging enhancer element
WO2014164697A1 (en) 2013-03-12 2014-10-09 Merial Limited Reverse genetics schmallenberg virus vaccine compositions, and methods of use thereof
GB201318804D0 (en) * 2013-10-24 2013-12-11 Adaptimmune Ltd Vectors for transgene expression
GB201322798D0 (en) 2013-12-20 2014-02-05 Oxford Biomedica Ltd Production system
NZ731659A (en) 2014-11-03 2018-10-26 Merial Inc Methods of using microneedle vaccine formulations to elicit in animals protective immunity against rabies virus
AU2016282772B2 (en) 2015-06-23 2019-09-05 Boehringer Ingelheim Animal Health USA Inc. PRRSV minor protein-containing recombinant viral vectors and methods of making and use thereof
CN116334143A (en) 2015-11-23 2023-06-27 诺华股份有限公司 Optimized lentiviral transfer vectors and uses thereof
US11427645B2 (en) 2017-03-15 2022-08-30 Oxford Biomedica (Uk) Limited 5T4-targeting agents and methods
ES2951857T3 (en) 2017-12-22 2023-10-25 Oxford Biomedica Ltd retroviral vector
CA3099497A1 (en) 2018-05-15 2019-11-21 Flagship Pioneering Innovations V, Inc. Fusosome compositions and uses thereof
AU2019301053A1 (en) 2018-07-09 2021-01-28 Flagship Pioneering Innovations V, Inc. Fusosome compositions and uses thereof
AU2019378883A1 (en) 2018-11-14 2021-06-03 Flagship Pioneering Innovations V, Inc. Fusosome compositions for T cell delivery
AU2019378881A1 (en) 2018-11-14 2021-06-03 Flagship Pioneering Innovations V, Inc. Fusosome compositions for CNS delivery
EP3880832A1 (en) 2018-11-14 2021-09-22 Flagship Pioneering Innovations V, Inc. Fusosome compositions for hematopoietic stem cell delivery
EP4025698A1 (en) 2019-09-03 2022-07-13 Sana Biotechnology, Inc. Cd24-associated particles and related methods and uses thereof
EP4058067A1 (en) 2019-11-12 2022-09-21 Oxford BioMedica (UK) Limited Production system
WO2021123832A1 (en) 2019-12-20 2021-06-24 Instil Bio (Uk) Limited Devices and methods for isolating tumor infiltrating lymphocytes and uses thereof
CN115667533A (en) 2020-02-13 2023-01-31 牛津生物医学(英国)有限公司 Production of lentiviral vectors
DE102020106710A1 (en) 2020-03-11 2021-09-16 Immatics US, Inc. WPRE MUTANT CONSTRUCTS, COMPOSITIONS, AND RELATED PROCEDURES
DE102020111571A1 (en) 2020-03-11 2021-09-16 Immatics US, Inc. WPRE MUTANT CONSTRUCTS, COMPOSITIONS, AND RELATED PROCEDURES
CN115667534A (en) 2020-03-13 2023-01-31 牛津生物医学(英国)有限公司 Lentiviral vectors
JP2023521663A (en) 2020-03-31 2023-05-25 サナ バイオテクノロジー,インコーポレイテッド TARGETED LIPID PARTICLES AND COMPOSITIONS AND USES THEREOF
CN116096430A (en) 2020-04-27 2023-05-09 衣阿华大学研究基金会 Compositions and methods for treating cystic fibrosis
GB202007199D0 (en) 2020-05-15 2020-07-01 Oxford Biomedica Ltd Viral vector production
US20230190871A1 (en) 2020-05-20 2023-06-22 Sana Biotechnology, Inc. Methods and compositions for treatment of viral infections
JP2023540705A (en) 2020-08-28 2023-09-26 サナ バイオテクノロジー,インコーポレイテッド Modified antiviral binding agents
GB202017725D0 (en) 2020-11-10 2020-12-23 Oxford Biomedica Ltd Method
JP2023554425A (en) 2020-12-18 2023-12-27 インスティル バイオ (ユーケイ) リミテッド Treatment of tumor-infiltrating lymphocytes
CA3205462A1 (en) 2020-12-18 2022-06-23 Instil Bio (Uk) Limited Processing of tumor infiltrating lymphocytes
EP4262828A1 (en) 2020-12-18 2023-10-25 Instil Bio (Uk) Limited Tumor infiltrating lymphocytes and anti-cd47 therapeutics
EP4274901A1 (en) 2021-01-11 2023-11-15 Sana Biotechnology, Inc. Use of cd8-targeted viral vectors
IL308836A (en) 2021-05-28 2024-01-01 Sana Biotechnology Inc Lipid particles containing a truncated baboon endogenous retrovirus (baev) envelope glycoprotein and related methods and uses
WO2023015217A1 (en) 2021-08-04 2023-02-09 Sana Biotechnology, Inc. Use of cd4-targeted viral vectors
WO2023038055A1 (en) * 2021-09-08 2023-03-16 タカラバイオ株式会社 Nucleic acid for retrovirus vector production
GB202114532D0 (en) 2021-10-12 2021-11-24 Oxford Biomedica Ltd Lentiviral Vectors
GB202114534D0 (en) 2021-10-12 2021-11-24 Oxford Biomedica Ltd Novel viral regulatory elements
GB202114528D0 (en) 2021-10-12 2021-11-24 Oxford Biomedica Ltd Lentiviral vectors
GB202114530D0 (en) 2021-10-12 2021-11-24 Oxford Biomedica Ltd Retroviral vectors
GB202114529D0 (en) 2021-10-12 2021-11-24 Oxford Biomedica Ltd Lentiviral vectors
WO2023077107A1 (en) 2021-10-29 2023-05-04 Sana Biotechnology, Inc. Methods and reagents for amplifying viral vector nucleic acid products
GB202117844D0 (en) 2021-12-09 2022-01-26 Oxford Biomedica Ltd Purification method
WO2023114949A1 (en) 2021-12-16 2023-06-22 Sana Biotechnology, Inc. Methods and systems of particle production
WO2023115039A2 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae fusion glycoproteins
TW202342757A (en) 2021-12-17 2023-11-01 美商薩那生物科技公司 Modified paramyxoviridae attachment glycoproteins
WO2023133595A2 (en) 2022-01-10 2023-07-13 Sana Biotechnology, Inc. Methods of ex vivo dosing and administration of lipid particles or viral vectors and related systems and uses
WO2023150518A1 (en) 2022-02-01 2023-08-10 Sana Biotechnology, Inc. Cd3-targeted lentiviral vectors and uses thereof
WO2023150647A1 (en) 2022-02-02 2023-08-10 Sana Biotechnology, Inc. Methods of repeat dosing and administration of lipid particles or viral vectors and related systems and uses
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies
WO2024026377A1 (en) 2022-07-27 2024-02-01 Sana Biotechnology, Inc. Methods of transduction using a viral vector and inhibitors of antiviral restriction factors
GB202211935D0 (en) 2022-08-16 2022-09-28 Oxford Biomedica Ltd envelope proteins
WO2024044655A1 (en) 2022-08-24 2024-02-29 Sana Biotechnology, Inc. Delivery of heterologous proteins
WO2024064838A1 (en) 2022-09-21 2024-03-28 Sana Biotechnology, Inc. Lipid particles comprising variant paramyxovirus attachment glycoproteins and uses thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5631154A (en) * 1988-06-10 1997-05-20 Therion Biologics, Incorporated Self assembled, defective, non-self-propagating lentivirus particles
US6013516A (en) * 1995-10-06 2000-01-11 The Salk Institute For Biological Studies Vector and method of use for nucleic acid delivery to non-dividing cells
US6207455B1 (en) * 1997-05-01 2001-03-27 Lung-Ji Chang Lentiviral vectors
US6300118B1 (en) * 1995-06-07 2001-10-09 American Home Products Corporation Plasmids comprising a genetically altered feline immunodeficiency virus genome
US6312682B1 (en) * 1996-10-17 2001-11-06 Oxford Biomedica Plc Retroviral vectors
US6428953B1 (en) * 1997-12-12 2002-08-06 Cell Genesys, Inc. Method and means for producing high titer, safe, recombinant lentivirus vectors
US6521457B2 (en) * 1997-05-13 2003-02-18 University Of North Carolina At Chapel Hill Lentivirus-based gene transfer vectors
US6555107B2 (en) * 1997-09-24 2003-04-29 The Regents Of The University Of California Lentiviral nucleic acids and uses thereof

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5747324A (en) * 1988-06-10 1998-05-05 Therion Biologics Corporation Self-assembled, defective, non-self-propagating lentivirus particles
JP3247972B2 (en) * 1990-05-28 2002-01-21 財団法人阪大微生物病研究会 A method for preparing a plasmid having both the ability to express a large amount of retrovirus gene and the processing ability after translation, and the plasmid obtained therefrom and its expression product
EP0536234A4 (en) 1990-06-20 1993-09-15 Dana Farber Cancer Institute Vectors containing hiv packaging sequences, packaging defective hiv vectors, and uses thereof
US5981276A (en) * 1990-06-20 1999-11-09 Dana-Farber Cancer Institute Vectors containing HIV packaging sequences, packaging defective HIV vectors, and uses thereof
WO1992021750A1 (en) 1991-05-29 1992-12-10 THE UNITED STATES OF AMERICA, represented by THE SCRETARY, DEPARTMENT OF HEALTH & HUMAN SERVICES Eukaryotic expression vectors with regulation of rna processing
EP0611822B1 (en) 1993-02-17 2002-05-22 Wisconsin Alumni Research Foundation More complex type retroviruses having mixed type LTR, and uses thereof
ATE381624T1 (en) * 1994-05-09 2008-01-15 Oxford Biomedica Ltd RETROVIRAL VECTORS WITH REDUCED RECOMBINATION RATE
EP0759471A4 (en) 1994-05-10 1997-10-15 Hisamitsu Pharmaceutical Co Recombinant human immunodeficiency virus vector and process for producing the same
CA2198210C (en) * 1994-09-02 2006-07-11 Walter Henry Gunzburg Non self-inactivating, expression targeted retroviral vectors
US6033672A (en) * 1996-03-15 2000-03-07 University Of Southern California Method of stimulating an immune response to caprine arthritis-encephalitis virus (CAEV) in humans through the administration of CAEV immunogens
JP2001513643A (en) * 1997-03-06 2001-09-04 ウーベルラ、クラウス Vectors and vector systems based on lentivirus
GB9720465D0 (en) * 1997-09-25 1997-11-26 Oxford Biomedica Ltd Dual-virus vectors
DK1895010T3 (en) * 1997-12-22 2011-11-21 Oxford Biomedica Ltd Vectors based on viruses for infectious horse anemia (eiav)

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5631154A (en) * 1988-06-10 1997-05-20 Therion Biologics, Incorporated Self assembled, defective, non-self-propagating lentivirus particles
US6300118B1 (en) * 1995-06-07 2001-10-09 American Home Products Corporation Plasmids comprising a genetically altered feline immunodeficiency virus genome
US6013516A (en) * 1995-10-06 2000-01-11 The Salk Institute For Biological Studies Vector and method of use for nucleic acid delivery to non-dividing cells
US6312682B1 (en) * 1996-10-17 2001-11-06 Oxford Biomedica Plc Retroviral vectors
US6207455B1 (en) * 1997-05-01 2001-03-27 Lung-Ji Chang Lentiviral vectors
US6521457B2 (en) * 1997-05-13 2003-02-18 University Of North Carolina At Chapel Hill Lentivirus-based gene transfer vectors
US6555107B2 (en) * 1997-09-24 2003-04-29 The Regents Of The University Of California Lentiviral nucleic acids and uses thereof
US6428953B1 (en) * 1997-12-12 2002-08-06 Cell Genesys, Inc. Method and means for producing high titer, safe, recombinant lentivirus vectors

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100003746A1 (en) * 2007-02-12 2010-01-07 Lesch Hanna P Production of Lentiviral Vectors
WO2022204351A3 (en) * 2021-03-24 2022-11-24 National Resilience, Inc. Systems and methods for cell-free polypeptide synthesis

Also Published As

Publication number Publication date
CN1285000A (en) 2001-02-21
AU760287B2 (en) 2003-05-08
WO1999032646A1 (en) 1999-07-01
JP4578678B2 (en) 2010-11-10
DK1895010T3 (en) 2011-11-21
CA2315269A1 (en) 1999-07-01
EP1895010B1 (en) 2011-10-12
EP1895010A2 (en) 2008-03-05
CN1322137C (en) 2007-06-20
US20060024274A1 (en) 2006-02-02
ES2373406T3 (en) 2012-02-03
EP1895010A3 (en) 2008-04-16
EP1042493A1 (en) 2000-10-11
ATE528405T1 (en) 2011-10-15
JP2002516065A (en) 2002-06-04
US6312683B1 (en) 2001-11-06
AU1773299A (en) 1999-07-12
CY1112196T1 (en) 2015-12-09
PT1895010E (en) 2012-01-25

Similar Documents

Publication Publication Date Title
US6312683B1 (en) Equine infectious anemia virus vectors
JP7340471B6 (en) Viral vector production system
AU2019203955B2 (en) Multipartite signaling proteins and uses thereof
CN111372943B (en) Adenovirus and use thereof
KR102319845B1 (en) CRISPR-CAS system for avian host cells
KR102196884B1 (en) Vectors for expression of prostate-associated antigens
US20040076613A1 (en) Vector system
KR102494564B1 (en) Malaria vaccine
AU2016317936A1 (en) Recombinant vectors comprising 2A peptide
CN114585366A (en) Cortical neural progenitor cells from ipscs
KR20220121844A (en) Compositions and methods for simultaneously regulating the expression of genes
US6544779B1 (en) Pseudo-type retroviral vectors with modifiable surface capsid proteins
KR20230019063A (en) Triple function adeno-associated virus (AAV) vectors for the treatment of C9ORF72 associated diseases
CN115927299A (en) Methods and compositions for increasing double-stranded RNA production
EP1859813A1 (en) Remedy for disease associated with apoptotic degeneration in ocular cell tissue with the use of siv-pedf vector
US20040071675A1 (en) Vector system
KR20230093326A (en) Chicken anemia virus (CAV)-based vectors
DK2921048T3 (en) SUS SCROFA V2G: SAFE HARBOR PLACE FOR LONG-TERM EXPRESSION AND HIGH INTEGRATION OF TRANSGENERS IN A PIG
KR20240024172A (en) Compositions and methods for regulating gene expression
GB2356200A (en) Hybrid viral vectors
WO2002094989A2 (en) Retroviral vectors and methods of using same
WO2000075351A1 (en) Siv-based packaging-defficient vectors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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