WO2000052188A1 - Cellules d'encapsidation pour vecteurs retroviraux - Google Patents

Cellules d'encapsidation pour vecteurs retroviraux Download PDF

Info

Publication number
WO2000052188A1
WO2000052188A1 PCT/GB2000/000766 GB0000766W WO0052188A1 WO 2000052188 A1 WO2000052188 A1 WO 2000052188A1 GB 0000766 W GB0000766 W GB 0000766W WO 0052188 A1 WO0052188 A1 WO 0052188A1
Authority
WO
WIPO (PCT)
Prior art keywords
retroviral
cell
expression
vsv
vector
Prior art date
Application number
PCT/GB2000/000766
Other languages
English (en)
Inventor
Yasuhiro Takeuchi
Mary Katharine Levinge Collins
Kyriacos Andreou Mitrophanous
Fiona Margaret Ellard
Susan Mary Kingsman
Original Assignee
Oxford Biomedica (Uk) Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Oxford Biomedica (Uk) Limited filed Critical Oxford Biomedica (Uk) Limited
Priority to AU29260/00A priority Critical patent/AU2926000A/en
Publication of WO2000052188A1 publication Critical patent/WO2000052188A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13051Methods of production or purification of viral material
    • C12N2740/13052Methods 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
    • C12N2740/15051Methods 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/108Plasmid DNA episomal 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/42Vector systems having a special element relevant for transcription being an intron or intervening sequence for splicing and/or stability of RNA
    • 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
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES

Definitions

  • the present invention relates to packaging and producer cell lines for producing recombinant viral vectors.
  • the present invention relates to methods for producing pseudotyped viral vectors with a broad host range which can be produced at in temperature regulated packaging and/or producer cells.
  • the invention relates to the generation of pseudotyped retroviral vectors, from stable producer cell lines, having vesicular stomatitis virus-G protein (VSV-G) as the membrane-associated viral envelope protein
  • VSV-G vesicular stomatitis virus-G protein
  • Retroviruses and vectors derived from them require an envelope protein in order to transduce efficiently a target cell.
  • the envelope protein is expressed in the cell producing the virus or vector and becomes incorporated into the virus or vector particles.
  • Retrovirus particles are composed of a proteinaceous core derived from the gag gene that encases the viral R A. The core is then encased in a portion of cell membrane that contains an envelope protein derived from the viral env gene.
  • the envelope protein is produced as a precursor, which is processed into two or three units. These are the surface protein (SU) which is completely external to the envelope, the transmembrane protein (TM) which interacts with the SU and contains a membrane spanning region and a cytoplasmic tail (Coffin 1992 In The Retroviridae, Pleum Press, ed Levy). In some retroviruses a small peptide is removed from the TM.
  • SU surface protein
  • TM transmembrane protein
  • the envelope protein In order to act as an effective envelope protein, capable of binding to a target cell surface and mediating viral entry, the envelope protein has to interact in a precise manner with the appropriate receptor or receptors on the target cell. This must occur in such a way as to result in internalisation of the viral particle in an appropriate manner to deliver the genome to the correct compartment of the cell to allow a productive infection to occur.
  • pseudotyping There have been many attempts to use the envelope protein derived from one virus to package a different virus, this is known as pseudotyping. The efficiency of pseudotyping is highly variable and appears to be strongly influenced by interactions between the cytoplasmic tail of the envelope and the core proteins of the viral particle.
  • visna virus can only be efficiently pseudotyped with HIN-1 envelope protein if the amino terminus of the matrix domain of the visna virus gag polyprotein is replaced by the equivalent HIN-1 matrix domain (Dorfman et al, 1994 J.
  • retroviral vectors In the construction of retroviral vectors it is desirable to engineer vectors with different target cell specificity's to the native virus, to enable the delivery of genetic material to an expanded or altered range of cell types.
  • One manner in which to achieve this is by engineering the virus envelope protein to alter its specificity.
  • Another approach is to introduce a heterologous envelope protein into the vector to replace or add to the native envelope protein of the virus.
  • the MLN envelope protein is capable of pseudotyping a variety of different retroviruses.
  • MLN envelope protein from an ampho tropic virus allows transduction of a broad range of cell types including human cells.
  • the envelope glycoprotein (G) of Nesicular stomatitis virus (NSN), a rhabdovirus, is another envelope protein that has been shown to be capable of pseudotyping certain retroviruses. Its ability to pseudotype MoMLN- based retroviral vectors in the absence of any retroviral envelope proteins was first shown by Emi et al (1991 Journal of Virology 65:1202-1207). WO94/294440 teaches that retroviral vectors may be successfully pseudotyped with NSN-G. These pseudotyped NSN-G vectors may be used to transduce a wide range of mammalian cells. Even more recently, Abe et al (J Nirol 1998 72(8) 6356-6361) teach that non-infectious retroviral particles can be made infectious by the addition of NSN-G.
  • NSN-G pseudotyped vectors have been shown to infect not only mammalian cells, but also cell lines derived from fish, reptiles and insects (Burns et al 1993 ibid). They have also been shown to be more efficient than traditional ampho tropic envelopes for a variety of cell lines (Yee et al, 1994 Proc. ⁇ atl. Acad. Sci.
  • NSN-G protein can be used to pseudotype certain retroviruses because its cytoplasmic tail is capable of interacting with the retroviral cores.
  • NSN-G protein pseudotyping envelope such as NSN-G protein gives the advantage that vector particles can be concentrated to a high titre without loss of infectivity (Akkina et al, 1996 J. Nirol. 70: 2581-5). Retrovirus envelope proteins are apparently unable to withstand the shearing forces during ultracentrifugation, probably because they consist of two non-covalently linked subunits. The interaction between the subunits may be disrupted by the centrifugation. In comparison the NSN glycoprotein is composed of a single unit. NSN-G protein pseudotyping can therefore offer potential advantages.
  • WO96/35454 teaches that a tetracycline responsive promoter may be used in combination with a nucleotide sequence enocoding vesicular stomatitis virus (NSN-G) to derive a retroviral packaging cell line that inducibly expresses the NSG-G protein at levels sufficient to support high level virus production but without the toxic effects of constitutive expression of NSN-G.
  • NSN-G nucleotide sequence enocoding vesicular stomatitis virus
  • Yoshida et al developed an adenovirus system to produce MoMLV vectors pseudotyped with VSV-G.
  • a cell line was produced containing a genome plasmid.
  • this cell line was infected with three different adenoviruses, one encoding the gag-pol gene of MoMLV under the control of the tetracycline transactivator, the second encoding VSV-G under the control of the tetracycline transactivator and the third encoding a nuclear localising transactivator. Transducing particles could be harvested from the resultant cells for a limited time period.
  • VSV glycoprotein mutants have used vaccinia virus systems in which the glycoprotein gene was cloned downstream of a bacteriophage T7 promoter. Co-infection of cells with the glycoprotein encoding vaccinia and a vaccinia virus expressing T7 polymerase resulted in a high level of expression of the VSV-G protein (Lefkowitz et al, 1990, Virology 178;373-383).
  • Arai et al reported the development of packaging cell lines in which a completely silent gene for the VSV glycoprotein was present to negate the above problem. This was achieved using a system in which a cassette was produced which encoded the CAG (the chicken ⁇ -actin gene promoter connected with the cytomegalo virus immediate-early promoter) followed by the 5' loxP sequence followed by the neo gene with an associated poly A signal followed by the 3' loxP sequence followed by the coding sequence for VSV-G and an associated poly A signal. When transfected into cells only the neo gene product is produced. If an adenovirus encoding the Cre recombinase is then introduced into the cell the neo sequence is removed by recombination and the VSV-G gene is expressed from the CAG promoter.
  • CAG the chicken ⁇ -actin gene promoter connected with the cytomegalo virus immediate-early promoter
  • Another problem associated with some of the systems described above is that chemicals need to be added to the culture medium to induce transducing particle production. These chemicals could potentially have deleterious side effects on the patient and so would require removal prior to formulation. Moreover, in some methods the cultures will be contaminated with proteins from other viruses, such as adenoviruses, which could activate unwanted immune responses when used to treat any patients.
  • the present invention seeks to overcome some of the problems associated with the prior art by providing stable cell lines, capable of producing transducing viral particles, that are capable of expressing VSV-G and which are produced without the use of added chemicals.
  • the present invention provides a packaging cell comprising: a first nucleotide sequence (NS) encoding a toxic viral envelope protein; and a second NS encoding a retrovirus nucleocapsid protein; wherein the expression of the first NS is regulatable in a temperature range from 25°C to 40°C.
  • NS nucleotide sequence
  • the temperature range is from 28 °C to 40°C.
  • the temperature range is from 32°C to 40°C.
  • the temperature range is from 35°C to 40°C.
  • the temperature range is from 28 °C to 37°C.
  • the temperature range is from 32°C to 37°C.
  • the temperature range is from about 32°C to 37°C.
  • the present invention provides a producer cell comprising: a first NS encoding a toxic viral envelope protein; a second NS encoding a retrovirus nucleocapsid protein; and a third NS comprising a retroviral sequence capable of being encapsidated in the nucleocapsid protein; wherein the retroviral vector particle titre obtained from same is dependent upon the temperature regulatable expression of the toxic viral envelope protein.
  • the present invention demonstrates that, despite the toxicity of VSV-G, it is possible to construct a stable retroviral producer line that is capable of producing transducing vector particles expressing VSV-G. Thus, the present invention demonstrates that the difficulty associated with producing stable cell lines capable of expressing VSV-G can be overcome.
  • the present invention is the first report of the propagation of stable cell lines that have the capacity to express high levels of the VSV-G protein.
  • the present invention also demonstrates the surprising finding that a VSV-G cell line produced using a pHCMV-G plasmid has a temperature sensitive phenotype and a VSV-G cell line produced using a pRV67 plasmid has a temperature insensitive phenotype.
  • This temperature sensitive VSV-G phenotype was characterised by the latent ability of a VSV-G cell line, transfected with the pHCMV-G plasmid, to produce significant quantities of a VSV-G protein at 32°C. Such a result has not been obtained before.
  • the surprising findings of the present invention are advantageous because they do not require the removal of chemicals or foreign viruses, such as adenovirus, from the viral particles produced.
  • Pseudotyping refers to a technique or strategy whereby an env gene is replaced with a heterologous env gene. Pseudotyping is not a new phenomenon and examples may be found in WO-A-98/05759, WO-A-98/05754, WO-A-97/17457, WO- A-96/09400, WO-A-91/00047 and Mebatsion et al 1997 Cell 90, 841-847.
  • heterologous refers to a nucleic acid sequence or protein sequence linked to a nucleic acid or protein sequence which it is not naturally linked.
  • variant in relation to this aspect of the present invention include any substitution of, variation of, modification of, replacement of, deletion of or addition of one (or more) nucleic acid (s) from or to the nucleotide sequence providing the expression product of the resultant nucleotide sequence has a temperature sensitive phenotype, preferably having at least the same temperature sensitive phenotype as the expression product of the sequence shown as SEQ ID No. 1 or SEQ ID No. 2.
  • Sequence identity with respect to SEQ ID No. 1 or SEQ ID No. 2 can be determined by a simple "eyeball” comparison (i.e. a strict comparison) of any one or more of the sequences with another sequence to see if that other sequence has, for example, at least 75% sequence identity to the sequence(s).
  • Relative sequence identity can also be determined by commercially available computer programs that can calculate % identity between two or more sequences using any suitable algorithm for determining identity, using for example default parameters.
  • a typical example of such a computer program is CLUSTAL.
  • the BLAST algorithm is employed, with parameters set to default values.
  • the BLAST algorithm is described in detail at http://www.ncbi.nih.gov/BLAST/blast_help.html, which is incorporated herein by reference.
  • the search parameters are defined as follows, can be advantageously set to the defined default parameters.
  • substantially identical when assessed by BLAST equates to sequences which match with an EXPECT value of at least about 7, preferably at least about 9 and most preferably 10 or more.
  • the default threshold for EXPECT in BLAST searching is usually 10.
  • BLAST Basic Local Alignment Search Tool
  • blastp, blastn, blastx, tblastn, and tblastx these programs ascribe significance to their findings using the statistical methods of Karlin and Altschul (see http://www.ncbi.nih.gov/BLAST/blast_help.html) with a few enhancements.
  • the BLAST programs were tailored for sequence similarity searching, for example to identify homologues to a query sequence. For a discussion of basic issues in similarity searching of sequence databases, see Altschul et al (1994) Nature Genetics 6:119-129.
  • the five BLAST programs available at http://www.ncbi.nlm.nih.gov perform the following tasks:
  • blastp - compares an amino acid query sequence against a protein sequence database.
  • blastx compares the six-frame conceptual translation products of a nucleotide query sequence (both strands) against a protein sequence database.
  • tblastn compares a protein query sequence against a nucleotide sequence database dynamically translated in all six reading frames (both strands).
  • tblastx compares the six-frame translations of a nucleotide query sequence against the six-frame translations of a nucleotide sequence database.
  • BLAST uses the following search parameters:
  • HISTOGRAM - Display a histogram of scores for each search; default is yes. (See parameter H in the BLAST Manual).
  • DESCRIPTIONS Restricts the number of short descriptions of matching sequences reported to the number specified; default limit is 100 descriptions. (See parameter V in the manual page).
  • EXPECT The statistical significance threshold for reporting matches against database sequences; the default value is 10, such that 10 matches are expected to be found merely by chance, according to the stochastic model of Karlin and Altschul (1990). If the statistical significance ascribed to a match is greater than the EXPECT threshold, the match will not be reported. Lower EXPECT thresholds are more stringent, leading to fewer chance matches being reported. Fractional values are acceptable. (See parameter E in the BLAST Manual). CUTOFF - Cutoff score for reporting high-scoring segment pairs. The default value is calculated from the EXPECT value (see above).
  • HSPs are reported for a database sequence only if the statistical significance ascribed to them is at least as high as would be ascribed to a lone HSP having a score equal to the CUTOFF value. Higher CUTOFF values are more stringent, leading to fewer chance matches being reported. (See parameter S in the BLAST Manual). Typically, significance thresholds can be more intuitively managed using EXPECT.
  • ALIGNMENTS Restricts database sequences to the number specified for which high-scoring segment pairs (HSPs) are reported; the default limit is 50. If more database sequences than this happen to satisfy the statistical significance threshold for reporting (see EXPECT and CUTOFF below), only the matches ascribed the greatest statistical significance are reported. (See parameter B in the BLAST Manual).
  • MATRIX - Specify an alternate scoring matrix for BLASTP, BLASTX, TBLASTN and TBLASTX.
  • the default matrix is BLOSUM62 (Henikoff & Henikoff, 1992).
  • the valid alternative choices include: PAM40, PAM120, PAM250 and IDENTITY.
  • No alternate scoring matrices are available for BLASTN; specifying the MATRIX directive in BLASTN requests returns an error response.
  • FILTER - Mask off segments of the query sequence that have low compositional complexity, as determined by the SEG program of Wootton & Federhen (1993) Computers and Chemistry 17:149-163, or segments consisting of short-periodicity internal repeats, as determined by the XNU program of Claverie & States (1993) Computers and Chemistry 17:191-201, or, for BLASTN, by the DUST program of Tatusov and Lipman (see http://www.ncbi.nlm.nih.gov). Filtering can eliminate statistically significant but biologically uninteresting reports from the blast output (e.g., hits against common acidic-, basic- or proline-rich regions), leaving the more biologically interesting regions of the query sequence available for specific matching against database sequences.
  • Filtering is only applied to the query sequence (or its translation products), not to database sequences. Default filtering is DUST for BLASTN, SEG for other programs.
  • NCBI-gi causes NCBI gi identifiers to be shown in the output, in addition to the accession and/or locus name.
  • sequence comparisons are conducted using the simple BLAST search algorithm provided at http://www.ncbi.nlm.nih.gov/BLAST.
  • no gap penalties are used when determining sequence identity.
  • the present invention also encompasses nucleotide sequences that are complementary to the sequences presented herein, or any fragment or derivative thereof. If the sequence is complementary to a fragment thereof then that sequence can be used as a probe to identify similar promoter sequences in other organisms.
  • the present invention also encompasses nucleotide sequences that are capable of hybridising to the sequences presented herein, or any fragment or derivative thereof.
  • Hybridization means a "process by which a strand of nucleic acid joins with a complementary strand through base pairing" (Coombs J (1994) Dictionary of Biotechnology, Stockton Press, New York NY) as well as the process of amplification as carried out in polymerase chain reaction technologies as described in Dieffenbach CW and GS Dveksler (1995, PCR Primer, a Laboratory Manual, Cold Spring Harbor Press, Plainview NY).
  • nucleotide sequences that are capable of hybridizing to the nucleotide sequences presented herein under conditions of intermediate to maximal stringency.
  • Hybridization conditions are based on the melting temperature (Tm) of the nucleic acid binding complex, as taught in Berger and Kimmel (1987, Guide to Molecular Cloning Techniques, Methods in Enzymology, Vol 152, Academic Press, San Diego CA), and confer a defined "stringency” as explained below.
  • Maximum stringency typically occurs at about Tm-5°C (5°C below the Tm of the probe); high stringency at about 5°C to 10°C below Tm; intermediate stringency at about 10°C to 20°C below Tm; and low stringency at about 20°C to 25 °C below Tm.
  • a maximum stringency hybridization can be used to identify or detect identical nucleotide sequences while an intermediate (or low) stringency hybridization can be used to identify or detect similar or related nucleotide sequences.
  • the present invention covers nucleotide sequences that can hybridise to the nucleotide sequences of the present invention under stringent conditions (e.g. 65°C and O.lxSSC).
  • the present invention also encompasses nucleotide sequences that are capable of hybridising to the sequences that are complementary to the sequences presented herein, or any fragment or derivative thereof.
  • the present invention encompasses nucleotide sequences that are complementary to sequences that are capable of hybridising to the sequence of the present invention. These types of nucleotide sequences are examples of variant nucleotide sequences.
  • the term “variant” encompasses sequences that are complementary to sequences that are capable of hydridising to the nucleotide sequences presented herein.
  • progeny virions bearing the genome of one virus encapsidated by the envelope protein of another.
  • toxic refers to an expression product, such as a VSV-G protein whose expression detrimentally affects the metabolism of the target cells, thus limiting the quantity of this expression product produced in such cells.
  • Target cell simply refers to a cell which a vector, whether native or targeted, is capable of infecting or transducing.
  • a "toxic-protein” refers to a protein which exhibits cytotoxic or cytostatic behaviour in cells.
  • Preferred assays for the measurement of cytotoxicity include the MTT assay which uses a non-radioactive format to quantitative viable cell number in proliferation assays (e.g. Promega Corp., Madison, WI) or 3 H incorporation into nascent DNA.
  • MTT assay which uses a non-radioactive format to quantitative viable cell number in proliferation assays (e.g. Promega Corp., Madison, WI) or 3 H incorporation into nascent DNA.
  • a number of other assay formats may likewise be utilised to evaluate various parameters associated with cell toxicity, including for example, cell viability (dye exclusion, cell counting) and cell metabolism (dye reduction) (see Cell Biology, A laboratory Handbook, ed. J.E. Celis, Academic Press) for further details and methods.
  • Other representative examples of toxic proteins include but are not limited to ⁇ interferon, interleukin-2
  • viral envelope protein refers to the protein embedded in the membrane which encapsulates the nucleocapsid and which protein is responsible for binding to and entry of the infectious virus into the target cell.
  • the viral envelope protein may also be a fusogenic protein.
  • fusogenic protein refers to glycoproteins which cause cells within a culture to fuse in a multinucleate syncytia. Representative examples of fusogenic proteins include VSV-G and Rabies G protein.
  • nucleocapsid refers to at least the group specific viral core proteins (gag) and the viral polymerase (pol) of a retrrovirus genome. These proteins encapsidate the retrovirus-packagable sequences and themselves are further surrounded by a membrane containing an envelope glycoprotein.
  • the term NOI i.e. nucleotide sequence of interest
  • the DNA sequence can be, for example, a synthetic DNA sequence, a recombinant DNA sequence (i.e. prepared by use of recombinant DNA techniques), a cDNA sequence or a partial genomic DNA sequence, including combinations thereof.
  • the DNA sequence need not be a coding region. If it is a coding region, it need not be an entire coding region.
  • the DNA sequence can be in a sense orientation or in an anti-sense orientation. Preferably, it is in a sense orientation.
  • the DNA is or comprises cDNA.
  • the NOI or NOIs may be under the expression control of an expression regulatory element, usually a promoter or a promoter and enhancer.
  • the enhancer and/or promoter may be preferentially active in a hypoxic or ischaemic or low glucose environment, such that the NOI is preferentially expressed in the particular tissues of interest, such as in the environment of a tumour, arthritic joint or other sites of ischaemia.
  • the enhancer element or other elements conferring regulated expression may be present in multiple copies.
  • the enhancer and/or promoter may be preferentially active in one or more specific cell types - such as any one or more of macrophages, endothelial cells or combinations thereof.
  • cell types such as any one or more of macrophages, endothelial cells or combinations thereof.
  • Further examples include respiratory airway epithelial cells, hepatocytes, muscle cells, cardiac myocytes, synoviocytes, primary mammary epithelial cess and post-mitotically terminally differentiated non-replicating cells such as macrophages neurons.
  • promoter is used in the normal sense of the art, e.g. an RNA polymerase binding site.
  • 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/or enhancer may be constitutively efficient, or may be tissue or temporally restricted in their activity.
  • tissue restricted promoters/enhancers are those which are highly active in tumour cells such as a promoter/enhancer from a MUCl 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 grp7S or a grp94 gene.
  • the alpha fetoprotein (AFP) promoter is also a tumour-specific promoter.
  • One preferred promoter-enhancer combination is a human cytomegalovirus (hCMV) major immediate early (MIE) promoter/enhancer combination.
  • the promoters of the present invention are tissue specific. That is, they are capable of driving transcription of a NOI or NOI(s) in one tissue while remaining largely “silent" in other tissue types.
  • tissue specific means a promoter which is not restricted in activity to a single tissue type but which nevertheless shows selectivity in that they may be active in one group of tissues and less active or silent in another group.
  • a desirable characteristic of the promoters of the present invention is that they posess a relatively low activity in the absence of activated hypoxia-regulated enhancer elements, even in the target tissue.
  • One means of achieving this is to use "silencer" elements which suppress the activity of a selected promoter in the absence of hypoxia.
  • the level of expression of an NOI or NOIs under the control of a particular promoter may be modulated by manipulating the promoter region. For example, different domains within a promoter region may possess different gene regulatory activities. The roles of these different regions are typically assessed using vector constructs having different variants of the promoter with specific regions deleted (that is, deletion analysis). This approach may be used to identify, for example, the smallest region capable of conferring tissue specificity or the smallest region conferring hypoxia sensitivity.
  • tissue specific promoters may be particularly advantageous in practising the present invention.
  • these promoters may be isolated as convenient restriction digestion fragments suitable for cloning in a selected vector.
  • promoter fragments may be isolated using the polymerase chain reaction. Cloning of the amplified fragments may be facilitated by incorporating restriction sites at the 5' end of the primers.
  • hypoxia means a condition under which a particular organ or tissue receives an inadequate supply of oxygen
  • suitable NOI 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).
  • 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, such as in one or more specific cell types.
  • Suitable NOIs for use in the invention in the treatment or prophylaxis of cancer include NOIs encoding proteins which: destroy the target cell (for example a ribosomal toxin), act as: tumour suppressors (such as wild-type p53); activators of anti-tumour immune mechanisms (such as cytokines, co-stimulatory molecules and immunoglobulins); inhibitors of angiogenesis; or which provide enhanced drug sensitivity (such as pro-drug activation enzymes); indirectly stimulate destruction of target cell by natural effector cells (for example, strong antigen to stimulate the immune system or convert a precursor substance to a toxic substance which destroys the target cell (for example a prodrug activating enzyme).
  • target cell for example a ribosomal toxin
  • tumour suppressors such as wild-type p53
  • activators of anti-tumour immune mechanisms such as cytokines, co-stimulatory molecules and immunoglobulins
  • inhibitors of angiogenesis or which provide enhanced drug sensitivity (such as
  • Encoded proteins could also destroy bystander tumour cells (for example with secreted antitumour antibody- ribosomal toxin fusion protein), indirectly stimulated destruction of bystander tumour cells (for example cytokines to stimulate the immune system or procoagulant proteins causing local vascular occlusion) or convert a precursor substance to a toxic substance which destroys bystander tumour cells (eg an enzyme which activates a prodrug to a diffusible drug).
  • bystander tumour cells for example with secreted antitumour antibody- ribosomal toxin fusion protein
  • indirectly stimulated destruction of bystander tumour cells for example cytokines to stimulate the immune system or procoagulant proteins causing local vascular occlusion
  • convert a precursor substance to a toxic substance which destroys bystander tumour cells eg an enzyme which activates a prodrug to a diffusible drug.
  • NOI(s) encoding antisense transcripts or ribozymes which interfere with expression of cellular genes for tumour persistence (for example against aberrant myc transcripts in Burkitts lymphoma or against bcr-abl transcripts in chronic myeloid leukemia.
  • tumour persistence for example against aberrant myc transcripts in Burkitts lymphoma or against bcr-abl transcripts in chronic myeloid leukemia.
  • Suitable NOIs for use in the treatment or prevention of ischaemic heart disease include
  • NOIs encoding plasminogen activators.
  • Suitable NOIs for the treatment or prevention of rheumatoid arthritis or cerebral malaria include genes encoding anti-inflammatory proteins, antibodies directed against tumour necrosis factor (TNF) alpha, and anti- adhesion molecules (such as antibody molecules or receptors specific for adhesion molecules).
  • TNF tumour necrosis factor
  • hypoxia regulatable therapeutic NOIs can be found in PCT/GB95/00322 (WO-A-9521927).
  • the expression products encoded by the NOIs may be proteins which are secreted from the cell. Alternatively the NOI expression products are not secreted and are active within the cell. In either event, it is preferred for the NOI expression product to demonstrate a bystander effector or a distant bystander effect; that is the production of the expression product in one cell leading to the killing of additional, related cells, either neighbouring or distant (e.g. metastatic), which possess a common phenotype.
  • the NOI or NOIs of the present invention may also comprise one or more cytokine- encoding NOIs.
  • Suitable cytokines and growth factors include but are not limited to: ApoE, Apo-SAA, BDNF, Cardiotrophin-1, EGF, ENA-78, Eotaxin, Eotaxin-2, Exodus-2, FGF-acidic, FGF-basic, fibroblast growth factor- 10 (Marshall 1998 Nature Biotechnology 16: 129).FLT3 ligand (Kimura et al (1997), Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF- ⁇ l, insulin, IFN- ⁇ , IGF-I, IGF-II, IL-l ⁇ , IL-l ⁇ , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 a.a.), IL-9, IL-10
  • a "vector” denotes 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.
  • vectors used in recombinant DNA techniques include plasmids, chromosomes, artificial chromosomes or viruses.
  • expression vector refers to an assembly which is capable of directing the expression of a nucleotide sequence of interest (NOI).
  • NOI expression vector must include a promoter which, when transcribed, is operably linked to the NOI, as well as a polyadenylation sequence.
  • both the promoter and the polyadenylation sequence are from a source which is heterologous to the helper elements, gag-pol and env.
  • the expression vectors described herein may be contained within a plasmid construct.
  • expression cassette refers to a recombinant DNA molecule containing a desired coding sequence and appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequence in a particular host organism. Nucleic acid sequences necessary for expression in eucaryotic cells usually include promoters, enhancers, and termination and polyadenylation signals. The cassette can be removed and inserted into a vector or plasmid as a single unit.
  • operably linked denotes a relationship between a regulatory region (typically a promoter element, but may include an enhancer element) and the coding region of a gene, whereby the transcription of the coding region is under the control of the regulatory region.
  • a regulatory region typically a promoter element, but may include an enhancer element
  • vector particle mean virus and virus-like particles that are capable of introducing a nucleic acid into a cell through a viral-like entry mechanism.
  • vector particles can, under certain circumstances, mediate the transfer of NOIs into the cells they infect.
  • a retrovirus is capable of reverse transcribing its genetic material into DNA and inco ⁇ orating this genetic material into a target cell's DNA upon transduction. Such cells are designated herein as "target cells”.
  • a vector particle includes the following components: a retrovirus nucleic acid, which may contain one or more NOIs, a nucleocapsid encapsidating the nucleic acid, the nucleocapsid comprising nucleocapsid protein of a retrovirus, and a membrane surrounding the nucleocapsid.
  • the heterologous NOI may be operably linked to a promoter and encode a protein that is expressible in a target cell.
  • such a heterologous NOI is capable of being expressed from the retrovirus genome either from endogenous retroviral promoters such as long terminal repeat (LTR) , or from a heterologous promoter to which the heterologous gene or sequence is operably linked.
  • LTR long terminal repeat
  • vector particles When the vector particles are used to transfer NOIs into cells which they transduce, such vector particles also designated “viral delivery systems” or “retroviral delivery systems".
  • Viral vectors including retroviral vectors, have been used to transfer NOIs efficiently by exploiting the viral transduction process. NOIs cloned into the retroviral genome can be delivered efficiently to cells susceptible to transduction by a retrovirus. Through other genetic manipulations, the replicative capacity of the retroviral genome can be destroyed. The vectors introduce new genetic material into a cell but are unable to replicate.
  • the vector of the present invention can be delivered by viral or non-viral techniques.
  • 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), multivalent cations such as spermine, cationic lipids or polylysine, 1, 2,-bis (oleoyloxy)-3-(trimethylammonio) propane (DOTAP)-cholesterol complexes (Wolff and Trubetskoy 1998 Nature Biotechnology 16: 421) and combinations thereof.
  • CFAs cationic facial amphiphiles
  • DOTAP 1, 2,-bis (oleoyloxy)-3-(trimethylammonio) propane
  • DOTAP 1, 2,-bis (oleoyloxy)-3-(trimethylammonio) propane
  • DOTAP 1, 2,-bis (oleoyloxy)-3-(trimethylammoni
  • Viral delivery systems include but are not limited to adenovirus vector, an adeno- associated viral (AAV) vector, a herpes viral vector, a retroviral vector, a lentiviral vector, or a baculoviral vector.
  • AAV adeno-associated viral
  • retroviruses include but are not limited to: murine leukemia virus (MLV), human immunodeficiency virus (HIV), equine infectious anaemia virus (EIAV), mouse mammary tumour virus (MMTV), Rous sarcoma virus (RSV), Fujinami sarcoma virus (FuSV), Moloney murine leukemia virus (Mo-MLV), FBR murine osteosarcoma virus (FBR MSV), Moloney murine sarcoma virus (Mo-MSV), Abelson murine leukemia virus (A-MLV), Avian myelocytomatosis virus-29 (MC29), and Avian erythroblastosis virus (AEV).
  • MMV murine leukemia virus
  • HCV human immunodeficiency virus
  • EIAV equine infectious anaemia virus
  • MMTV mouse mammary tumour virus
  • RSV Rous sarcoma virus
  • FuSV Fujinami sarcoma virus
  • Preferred vectors for use in accordance with the present invention are recombinant viral vectors, in particular recombinant retroviral vectors (RRV).
  • the lentiviruses can be divided into primate and non-primate groups. Examples of primate lentiviruses include but are not limited to: the human immunodeficiency virus (HIV), the causative agent of human auto-immunodeficiency syndrome (AIDS), and the simian immunodeficiency virus (SIV).
  • the non-primate lentiviral group includes the prototype "slow virus” visna/maedi virus (VMN), as well as the related caprine arthritis-encephalitis virus (CAEV), equine infectious anaemia virus (EIAV) and the more recently described feline immunodeficiency virus (FIV) and bovine immunodeficiency virus (BIV).
  • VNN visna/maedi virus
  • CAEV caprine arthritis-encephalitis virus
  • EIAV equine infectious anaemia virus
  • FV feline immunodeficiency virus
  • BIV bovine immunodeficiency virus
  • lentiviruses have the capability to infect both dividing and non-dividing cells (Lewis et al 1992 EMBO. J 11 : 3053-3058; Lewis and Emerman 1994 J. Virol. 68: 510-516).
  • other retroviruses - such as MLV - are unable to infect non-dividing cells such as those that make up, for example, muscle, brain, lung and liver tissue.
  • Preferred vectors for use in accordance with the present invention are recombinant viral vectors, in particular recombinant lentiviral vectors.
  • vectors include ex vivo delivery systems, which include but are not limited to D ⁇ A transfection methods such as electroporation, D ⁇ A biolistics, lipid- mediated transfection, compacted D ⁇ A-mediated transfection.
  • D ⁇ A transfection methods such as electroporation, D ⁇ A biolistics, lipid- mediated transfection, compacted D ⁇ A-mediated transfection.
  • the vector may be a plasmid D ⁇ A vector.
  • the vector may be a recombinant viral vector.
  • Suitable recombinant viral vectors include adenovirus vectors, adeno-associated viral (AAV) vectors, Herpes-virus vectors, or retroviral vectors, lentiviral vectors or a combination of adenoviral and lentiviral vectors.
  • AAV adeno-associated viral
  • retroviral vectors lentiviral vectors or a combination of adenoviral and lentiviral vectors.
  • gene delivery is mediated by viral infection of a target cell.
  • the vector of the present invention may be configured as a split-intron vector.
  • a split intron vector is described in PCT patent application GB98/02885 and GB/98/02867. If the features of adenoviruses are combined with the genetic stability of retro/lentiviruses then essentially the adenovirus can be used to transduce target cells to become transient retroviral producer cells that could stably infect neighbouring cells.
  • a "packaging cell” refers to a cell which contains those elements necessary for production of infectious recombinant virus which are lacking in a recombinant viral vector.
  • packaging cells typically contain one or more expression cassettes which are capable of expressing viral structural proteins (such as gag, pol and env) but they do not contain a packaging signal.
  • packetaging signal which is refered to interchangeably as “packaging sequence” or “psi” is used in reference to the non-coding sequence required for encapsidation of retroviral RNA strands during viral particle formation.
  • Packaging cell lines suitable for use with the above-described vector constructs may be readily prepared (see also WO 92/05266), and utilised to create producer cell lines for the production of recombinant vector particles.
  • a “producer cell” or “vector producing cell” refers to a cell which contains all the elements necessary for production of recombinant viral vector particles and retroviral delivery systems.
  • the producer cell is obtainable from a stable producer cell line.
  • the producer cell is obtainable from a stable producer cell line constructed using pHCMV-G and a second plasmid with a selectable marker.
  • the toxic envelope protein sequences, and nucleocapsid sequences are all stably integrated in the cell.
  • one or more of these sequences could also exist in episomal form and gene expression could occur from the episome.
  • retroviral sequences are capable of being packaged with the nucleocapsid proteins.
  • the retroviral sequences that are capable of being packaged may also contain one or more heterologous NOIs that are capable of being expressed in a target cell that is infected by the virions produced in the producer cell.
  • the packaging cell lines are useful for providing the gene products necessary to encapsidate and provide a membrane protein for a retrovirus and retrovirus nucleic gene delivery vehicle.
  • retrovirus sequences are introduced into the packaging cell lines, such sequences are encapsidated with the nucleocapsid proteins and these units then bud through the cell membrane to become surrounded in cell membrane and to contain the envelope protein produced in the packaging cell line.
  • infectious retroviruses are useful as infectious units per se or as gene delivery vectors.
  • the cells are useful for producing infectious pseudotyped retrovirus, and especially high titer virions which may also contain one or more NOIs sequences capable of being expressed in a target cell or tissue.
  • the cells are thus useful for packaging a retrovirus genome which may also contain a heterologous NOI capable of being expressed in a target cell or tissue
  • Our invention is based on an unexpected observation made when constructing producer cell lines by introducing the plasmid pHCMV-G. This is the same plasmid as was used by Chen et al (1996) and Yee et al (1994) to express the VSV-G protein. Clonal cultures were selected and about 70% failed produce a detectable virus titre, the remaining 30% produced a variable amount of virus. It was observed that lowering the temperature of the incubator from 37°C to 32°C for a four day period increased the titre per cell by 30 fold. An increase in the surface expression of VSV-G at 32°C in comparison to 37°C was detected using monoclonal antibodies (specific to the cytoplasmic tail of VSV-G) and polyclonal antibodies (raised against the whole protein).
  • pHCMV-G pHCMV-G was introduced into either TE671 or HT1080 cells found that in the majority if not all of the cell lines found to express VSV-G, expression was low at 37°C and increased significantly at 32°C. At present no cell lines have been obtained with this plasmid, which show high levels of VSV-G expression at 37°C. In contrast, cell lines which produced high levels of VSV-G at 37°C could be obtained when another plasmid, pRV67, was used. So far, the increase in expression at 32°C in comparison to 37°C has not been observed in the cell lines constructed using pRV67 that expresses a fully functional variant of VSV-G.
  • Virions of rhabdoviruses are enveloped; protruding through the envelope is a homotrimeric membrane glycoprotein, G.
  • G This protein is responsible for the attachment of the virus to cellular receptors and is required for virus infectivity.
  • the glycoprotein of VSV serotype Indiana has 51 1 amino acids including a signal peptide, a hydrophobic membrane spanning domain of 20 amino acids, and a 29 amino acid hydrophilic carboxy-terminal cytoplasmic domain and usually has two asparagine- linked complex oligosaccharides attached (Rose and Gallione 1981 Journal of Virology 39: 519-528).
  • a fatty acid chain (palmitate) is linked to the single cysteine residue in the cytoplasmic domain (Rose et al, 1984 Proceedings of the National Academy of Science USA 81 : 2050-2054).
  • the protein is then transported to the Golgi apparatus and the oligosaccharides are further processed. This involves the removal of some glucose and mannose residues and the addition of the peripheral sugars, N-acetlyglucosamine, galactose, sialic acid and fructose.
  • the protein is then transported to the plasma membrane and is incorporated into virus particles.
  • Transport of the unglycosylated San Juan protein could be increased to similar levels to those observed in Orsay by mutating residue 172 to an aspartic acid (tyrosine in San Juan and aspartic acid in Orsay).
  • a measurable increase in the amount of glycoprotein transport in San Juan was also observed if residue 231 was mutated from a glycine to an aspartic acid.
  • G proteins in which one or both of the two glycosylation site have been mutated, are lower than in the wt constructs for both the San Juan and Orsay strains (Pitta et al, 1989, Journal of Virology 63: 3801-3809; Machamer et al, 1985, Molecular and Cellular Biology 5: 3074-3083). This did not appear to be due to increased degradation. It is therefore possible that the expression level of VSV-G might be lower in pRV67 than in pHCMV-G.
  • Figure 1 which shows a diagrammatic representation of VSV-G expression cassettes - pRV67 and pHCMV-G;
  • Figure 2 which shows a photographic representation of western blots showing the expression of the VSV glycoprotein in cytoplasmic extracts of various cell lines;
  • Figure 4 which shows a photographic representation of VSV-G expression in stable cloned cell lines derived from cell transfected with pHCMV-G;
  • Figure 5 which shows a photographic representation of the increase in VSV-G expression with the time of culture at 32°C;
  • Figure 6 which shows a diagrammatic representation of pONY2.1nlsZ ⁇ cZ and pONY3;
  • Figure 7 which shows a graphical representation that pRV67 derived cell lines do not show temperature regulated VSV-G expression.
  • Figure 8 which shows a graphical representationn of transient vector production in a PI cell line after different times at 32°C;
  • Figure 9 which shows a graphical representation of transient vector production in PI cells transfected 12 hours prior to shift down to 32°C;
  • Figure 10 shows a graphical representation of the effect of temperature on vector production from stable cell lines
  • Figure 1 1 which shows a photographic representation of the effect of temperature on VSV-G expression in a VSV671 cell line;
  • Figure 12 which shows a pictorial representation of MLV plasmids
  • Figure 13 which shows the production of MLV vector particles by the PI cell line
  • Figure 14 which presents a Northern blot analysis showing the increase in VSV-G mRNA levels at 32°C;
  • Figure 15 which shows a photographic representation of a western blot performed to screen clones obtained from transfecting VSV-G expressing cells with CeB for MLV p30 expression.
  • Figure 16a which shows the effects of temperature on particle production in a packaging cell line, PI -CeB clone 12 transduced with MLV vector particles.
  • Figure 16b which shows the effects of temperature on particle production in a packaging cell line PI -CeB clone 7 transduced with MLV vector particles.
  • Figure 17 which shows the effect of temperature on particle production in an MLV producer cell line, derived from P 1.
  • Figure 18 which shows details of the EIAV gag/pol expression plasmid, pONYHYG, used to produce packaging cell lines.
  • Figure 19 which shows a photographic representation of a western blot performed to screen clones for EIAV gag/pol expression obtained from transfecting VSV-G expressing cells with pONYHYG.
  • Figure 20 which shows a photographic representation of a western blot performed to screen clones for EIAV gag/pol expression obtained from limit diluting the ring clones AC4 and BD7.
  • FIG. 21 which shows the effect of sodium butyrate on transducing particle production in an EIAV producer cell line.
  • Figure 22 which shows a photographic representation of a southern blot of chromosomal DNA samples isolated from a range of cell lines restricted with three different enzymes, and probed with a VSV-G probe.
  • Figure 23 which shows a photographic representation of a southern blot of chromosomal DNA samples isolated from a range of cell lines restricted with three different enzymes, and probed with an EIAV gag/pol probe.
  • Figure 24 which shows a photographic representation of a northern blot of total RNA samples isolated from a range of cell lines and probed with a VSV-G probe.
  • Figure 25 which shows the results on an analysis of the increase in VSV-G mRNA levels in cells grown at 32 ° C compared to the levels observed at 37° C.
  • Figure 26 which shows a photographic representation of a western blot analysis of the increase in the level of VSV-G protein in cells grown at 32° C compared to cells grown at 37° C.
  • Figure 27 which shows a photographic representation of a northern blot of total RNA samples isolated from a range of cell lines and probed with a probe to EIAV gag/pol.
  • Figure 28 which shows the results on an analysis of the increase in EIAV gag/pol mRNA levels in cells grown at 32°C compared to the levels observed at 37 °C.
  • Figure 29 which presents SEQ ID No. 1;
  • Figure 1 shows details of the two plasmids used to express the VSV glycoprotein.
  • Figure 2 shows western blots illustrating the expression of the VSV glycoprotein in cytoplasmic extracts of various cell lines.
  • the protein was detected using the monoclonal antibody, P5D4, which binds to the cytoplasmic tail of the glycoprotein.
  • the cells were transiently transfected with the plasmids and analysed for expression after 48hrs at 37°C.
  • Figure 3 shows the one coding difference detected between the VSV-G genes in pHCMV-G and pRV67.
  • the sequence shown is from amino acid 333 to 340 (the numbering assumes that amino acid 1 is the first residue in the signal peptide sequence).
  • Figure 4 shows an example of the results obtained when cytoplasmic extracts of cloned cell lines derived from cell transfected with pHCMV-G were analysed for VSV-G expression.
  • Samples of the clones were grown at 37°C and 32°C for 3 days prior to harvesting and samples for both temperatures are shown.
  • VSV-G expression was detected using Western blotting and the monoclonal antibody (P5D4) as described in Example 1.
  • O samples are clones derived from HT1080 cells transfected with pHCMV-G and pCI-NEO and selected on lug/ml G418.
  • P samples are clones derived from TE671 cells transfected with pHCMV-G and pCI-NEO and selected on lug/ml G418.
  • Figure 5 shows the time required for VSV-G expression to increase when a cloned cell line, PI, derived from TE671 cells transfected with pHCMV-G was shifted to 32°C.
  • VSV-G expression was detected using Western blotting and the monoclonal antibody (P5D4) as described in Example 1.
  • Panel A shows an SDS-PAGE gel with ⁇ - mercapto-ethanol; and
  • Panel B shows a SDS-PAGE page without ⁇ -mercapto-ethanol.
  • Lanes numbered 1-7 and M contain the following samples:
  • Figure 6 shows details of the EIAV plasmids used to produce transducing particles from cell lines expressing VSV-G.
  • Figure 7 shows the results from experiments in which a cell line that expressed VSV- G, (A7) derived from pRV67, was transfected with pONY2.1nlsZ ⁇ cZ and pONY3. The medium from such transfected cells was removed every day and the number of transducing particles present per ml was determined using D17 cells.
  • Figure 8 shows a study to determine the optimum conditions for analysing vector production after transient transfection of gag-pol and genome into VSV-G pseudotyping cell lines.
  • Figure 9 shows results from a similar study to that in Figure 9 using PI cells transfected with pONY2.1nlsZ ⁇ cZ and pONY3. In this case the cells were shifted from 37°C to 32°C twelve hours after transfection. The medium from such transfected cells was removed every day and the number of transducing particles present per ml was determined on D17 cells.
  • Figure 10 shows the time course of production of MFG retroviral vector from TelCEB ⁇ lines containing HCMV-G. Three clones were analysed. They were grown at 37°C for 2 days and then shifted to 32°C. Culture supernatants were analysed by titration on D17 cell. Cell numbers were counted at each harvest point and data for VSV-7 cell line are shown.
  • Figure 11 shows HCMV-G expression from a typical TelCEB ⁇ cell line containing pHCMV-G (VSV671).
  • the cell line VSV671 was grown at 32°C (lane3) and 37°C.
  • Figure 12 shows plasmids used to produce MLV vectors.
  • Figure 13 shows the effect of reducing the temperature on the production of transducing particles from producer cell lines derived from TelCeB ⁇ cells transfected with pHCMV-G.
  • Figure 14 represents a northern blot of total RNA isolated from PI cells grown for different amounts of time at 32°C. The blot was probed with the complete coding region of the VSV-G gene. Lanes numbered 1-7 contain the following samples: 1 TelCeB cells grown at 37°C. 2 PI cells grown at 37° C.
  • VSV-G gene from pHCMV-G present on a 1692bp BamHl fragment.
  • Figure 29 presents SEQ ID No 1 which is the BamHl fragment containing the VSV-G gene present in pHCMV-G.
  • Figure 30 presents SEQ ID No 2 which is the BamHl fragment cloned into pSA91 to construct pRV67 which contains the VSV-G gene.
  • Plasmids that express the VSV-G protein and that are widely used for pseudotyping both retroviral and lentiviral vectors are shown in Figure 1. They were constructed as follows: pHCMV-G was constructed by Yee et al (1994 Proc. Natl. Acad. Sci. USA 91 : 9564-9568). The BamHl fragment containing the VSV-G gene was isolated from pLGRNL (Emi et al, 1991 Journal of Virology 65:1202-1207) and was inserted into the unique BamHl site in pHCMV-Bam. pHCMV-Bam contains the human cytomegalovirus immediate early promoter followed by the splicing and polyadenylation signals derived from the rabbit ⁇ -globin gene.
  • pRV67 was constructed by cloning a BamHl fragment containing the VSV-G gene into pSA91.
  • pSA91 is a derivative of pGWIHG (Soneoka et al 1995 Nucl. Acids Res.
  • VSV-G The expression of VSV-G from these plasmids is confirmed by western blot analysis of transiently transfected cells as shown in Figure 2.
  • the two plasmids were transfected into HT1080 (ATCC CCL 121) and TE671 (ATCC 8805-CRL) using FuGENE 6TM (Boehringer Mannheim). 48 hours after transfection the cells were rinsed in phosphate buffered saline (PBS) and were lysed in with 1% v/v Nonidet P40 in PBS. The protein concentrations of the samples were measured using the BioRad DC protein assay as per the manufacturer's instructions and 5 ⁇ g samples were analysed by SDS/PAGE followed by western blotting.
  • PBS phosphate buffered saline
  • VSV-G protein was detected using a mouse monoclonal antibody raised against a peptide (YTDIEMNRLGK) in the cytoplasmic tail of VSV-G (P5D4), the antibody is conjugated to horse radish peroxidase (Boehringer Mannheim) and can be used directly which ECL detection reagents. Cells were cultured at the usual temperature of 37°C.
  • pHCMV-G and pCl-neo Two different approaches were used to produce cell lines expressing VSV-G. pHCMV-G and pCl-neo (Promega) were transfected into HT1080 and TE671 using FuGENE 6TM (Boehringer Mannheim) at a ratio of 10 to 1.
  • FuGENE 6TM Boehringer Mannheim
  • pCI-oeo encodes for the aminoglycoside phosphotransferase gene down stream of an SV40 promoter, cells in which this gene is expressed should be resistant to the antibiotic Geneticin (Schering Corporation). The cells were then selected using lmg ml "1 Geneticin, the assumption being that the majority of the cells that contained pCl-neo would also contain pHCMV-G.
  • Cytoplasmic extracts from the clones were analysed for the presence of VSV-G protein using western blotting and the monoclonal antibody (P5D4) as described in Example 1.
  • Samples of the clones were grown at 37°C and also at 32°C.
  • An example of the results obtained with cells engineered to contain pHCMV-G is shown in Figure 4. They reveal the surprising result that the expression levels of VSV-G are dramatically reduced at 37°C as compared to 32°C. There was variability between the clones in terms of the maximum level of expression but even in this small sample clone 022 and clone PI were obtained that gave a strong temperature dependent control of VSV-G expression.
  • VSV-G in cell lines derived from pHCMV-G when the culture temperature is shifted from 37°C to 32°C was characterised further. Monolayers of cells were grown in six well tissue culture dishes at 37°C until 50% confluent at which time the plates were shifted to 32°C. Cells were harvested every day as in Example 1 and the amount of VSV-G present was analysed using western blotting. Samples were detected using the monoclonal antibody P5D4 to VSV-G. The results are shown in Figure 5.
  • the VSV-G protein can be seen migrating just in front of a cross reacting protein.
  • This upper protein is present even in lane 7 that contains the parental TE671 cell line.
  • the lower protein band is clearly absent from this lane confirming that this is indeed the VSV-G protein.
  • panel B the samples are run in their non-reduced form. That is, they are not treated with a reducing agent that breaks cysteine bonds in the protein. In this case there are no proteins detected in the TE671 negative control sample (see panel B) as presumably the cross reactive epitope on the cellular protein is not revealed without reduction.
  • VSV-G expressing cell lines could be used to produce lentiviral vectors.
  • the assay system is based on the three-plasmid transfection method described previously (Soneoka et al, 1995 ibid).
  • the plasmids used in these experiments were as follows; a genome plasmid, pONY2.1nlsZ ⁇ cZ or a derivative of it, and a plasmid that expresses the EIAV gag-pol genes pONY3 or a derivative of it (GB patent application 9727135.7). The important features of these vectors are shown in Figure 6.
  • EIAV vectors were as follows. An infectious proviral clone, pSPEIAV19, as described by Payne et al (1994, J.Gen. Virol. 75:425-9) was used as a starting point. A plasmid, pSPEIAV ⁇ H, was constructed by the deletion of a Hindlll fragment, 5835-6571, from the region of the plasmid encoding for the envelope protein. A vector genome plasmid was constructed by inserting the EIAV LTR, amplified by PCR from pSPEIAV19, into pBluescript II KS+ (Stratagene).
  • the MluVMlul (216/814) fragment of pSPEIAV ⁇ H was then inserted into the LTR/Bluescript plasmid to generate pONY2.
  • a BglU/Ncol fragment within pol (1901/4949) was deleted and a nuclear localising ⁇ -galactosidase gene driven by the HCMV IE enhancer/promoter was inserted in its place. This was designated pONY2.1nlsZ ⁇ cZ.
  • EIAV plasmid (pONY3) encoding the gag-pol genes was then made by inserting the MluVMlul fragment from pONY2 into the mammalian expression plasmid pCl-neo (Promega) such that the gag-pol protein is expressed from the HCMV IE enhancer/promoter.
  • pHCMV-G pHCMV-G
  • a temperature shift to 32°C is critical for generating vector.
  • the timing of the temperature shift relative to the transfection was critical and titres were dependent upon transfection efficiency.
  • stable producer lines were developed in which all of the components were stably maintained in the cell.
  • a cell line that produced transducing particles based on MLV was constructed.
  • TELCeB ⁇ a cell line that had already been produced to express the MLV gag-pol gene and a vector genome.
  • This cell line is derived from TE671 cells and harbours the MFGnls/ cZ retroviral vector and a MoMLV gag-pol expression plasmid (CeB), (Cosset et al 1995 Journal of Virology 69:7430-7436).
  • Flasks of cells were grown at 37°C until confluent and then were shifted to 32°C. A typical time course of virus production is shown in Figure 10. All three clones showed an increase in titre for seven days after the temperature shift. This increase was not simply due to an increase in the number of cells in culture which is shown for the VSV-7 cell line. After seven days at 32°C, the cells began to round up and detach and the yield of transducing particles decreased. An increase in the surface expression of VSV-G was detected using a monoclonal antibody (P5D4 specific to the cytoplasmic tail of VSV-G) and polyclonal antibodies (raised against the whole protein) at 32°C in comparison to 37°C.
  • P5D4 specific to the cytoplasmic tail of VSV-G
  • FIG. 11 shows the results of a Western blot performed on cell lysates from one of the stable producer cell lines developed. VSV-G expression was detected using the monoclonal antibody, P5D4. The presence of VSV-G can clearly be seen at 32°C, but not at 37°C.
  • the P 1 cell line described in example 2 which had been grown at 37°C for different periods of time (70 or 106 days after transfection with pHCMV-G) was transfected with the plasmids pMLV-GP (genome) and pCIE-GPSD (gag-pol).
  • the plasmid pMLV-GP (see Figure 12) was constructed by M. Yap (University of Oxford). pHITl l l (Soneoka et al, 1995 ibid) was restricted with Kpnl to remove the region between the LTRs, this was replaced by a similar Kpnl fragment from pLNCX (Miller and Rosman 1989, Biotechniques 7: 980-990).
  • the resultant plasmid was cut with Hpal and the enhanced green fluorescent protein gene (egfp) from pEGFP-Nl (Clontech) was inserted as a Smal-Hpal fragment.
  • pCIE-GPSD was constructed by inserting the gag/pol genes from MLV (derived from pHIT60; Soneoka et al, 1995 ibid) into a derivative of pCI- «e ⁇ (Promega) with an extended enhancer region.
  • the dihydrofolate reductase gene (dhfr) from pSV2-DHFR was inserted after the gag-pol genes with a short spacer of 74 residues separating the genes, this should allow some of the ribosomes which translate the gag-pol genes to reinitiated and translate the dhfr gene.
  • RNA was prepared from the PI cell line and analysed by northern blotting using a probe homologous to VSV-G mRNA ( Figure 14).
  • Figure 14 presents a Northern blot analysis showing an increase in VSV-G mRNA levels at 32°C.
  • PI a stable cell line
  • PI derived from TE671 cells transfected with pHCMV-G
  • time after the cells were shifted from 37 °C to 32°C Only small amounts of the protein were detected at 37°C, whilst significant amounts of the protein were found to be present after 3 days at 32°C. Since this increase could be due an effect of temperature on transcription or translation an experiment was performed to investigate whether VSV-G RNA levels were affected by changes in culture temperature.
  • PI cells were transferred from 37 °C to 32°C and cells were harvested every day for four days.
  • RNA content of the cells was isolated using TRIZOL reagent (GIBCOBRL) and a northern blot was performed using standard methods. This blot was probed using a 1.6kb DNA fragment containing the whole of the VSV-G.
  • GIBCOBRL TRIZOL reagent
  • the concepts and design body are broadly applicable to cell lines for the production of any viral vector where harmful or otherwise undesirable viral proteins must be produced by the cell in order for the viral vector to be produced.
  • the constructs and methods of invention are used to prevent or minimise the production of these proteins until they are needed. At that time, expression is induced for a period of time necessary for the production of the proteins and the assembly of the viral vectors.
  • viral vectors examples include but are not limited to other RNA viral vectors besides retroviral vectors, and DNA viral vectors, such as adenoviral vectors, adeno-associated viral vectors, Herpesvirus vectors (preferably Herpes simplex I virus vectors), and vaccinia virus vectors.
  • adenoviral vectors examples include, for adenoviral vectors, products of the El, E2, E4, and major late genes; for adeno-associated viruses, the rep protein; and for Herpesvirus, the capsid protein.
  • VSV-G a retroviral producer line that releases vector particles carrying VSV-G.
  • the method that we have developed involves passage of VSV-G expressing cell lines at 37°C and then screening cells for enhanced production at 32°C.
  • this effect may be dependent upon the integrity of the second glycosylation site in VSV-G as found in plasmid pHCMV-G. Mutation of this site in pRV67 although not influencing the ability of the protein to pseudotype vectors does not allow the selection of a temperature regulated producer clone although surprisingly there was sufficient expression at 37°C to allow some vector production.
  • PI cells were transfected using FuGENE6TM (Roche) with CeB (Cosset et al. 1995) and 14 clones were selected with 3 ⁇ g per ml blasticidin.
  • the MLV gag/pol expression levels of these clones was determined using western blotting ( Figure 15) using a monoclonal antibody R187 (Chesebro et al.127: 134-148) to MLV p30 gag, 15 ⁇ g of total protein from cell lysates was loaded in each lane of the gel. These expression levels were compared with the expression levels found in two MLV based packaging cell lines, FLYA13 (Cosset et al.
  • MLV particles were produced using the three-plasmid transfection method described previously (Soneoka et al, 1995).
  • the plasmids used in the transfection were as follows; a genome plasmid, pMLV-GP ( Figure 12), pHIT60 (MLV gag/pol) and pHIT456 (MLV amphotropic envelope; the latter plasmids are as Soneoka et al. (1995).
  • the resulting vector particles were used to transduce PI -CeB clones 7 and 12.
  • the transduced cells were grown at 37°C for seven days, the cells were then transferred to six well plates and were cultured at either 37°C or 32°C.
  • PI cells were transfected using FuGENE ⁇ TM (Roche) with pONYHYG.
  • This plasmid (see Figure 18) is a derivative of pONY3 (GB patent application 9727135.7) in which an antibiotic resistance marker is linked to the expression of EIAV gag/pol using an IRES (internal ribosome entry site).
  • IRES internal ribosome entry site.
  • Potential EIAV gag/pol expressing cell lines were selected using 200 to 400 ⁇ M hygromycin B (Roche). Seven clones were selected and the amount of gag/pol expressed by each clone was quantified using western blotting.
  • EIAV protein was detected using a polyclonal serum from an EIAV infected horse and ECL.
  • Four of the seven clones were shown to express detectable amounts of gag/pol (see Figure 19). However it is clear that in order to produce a commercially viable packaging cell line, typically more than seven clones would be assessed. In this study, stable cell lines were used in which the levels of gag/pol expression are far higher than those observed for the present clones.
  • Two clones AC4 and BD7 were limit dilute cloned to ensure clonality and were reassessed for the levels of gag/pol that they expressed ( Figure 20). Two clones were selected for further study, AC4 clone 4 (named EV2) and BD7 clone 2 (named EV1).
  • gag/pol expression may be achieved in both EIAV and MLV producer cell lines and that genome expression levels are not optimal in the EIAV model.
  • commercially useful producer cell lines can be developed using a system in which VSV-G expression is regulated by temperature.
  • Hindlll does not cut within the coding sequence and therefore may provide information on how many copies in the VSV-G gene have integrated into the genomes of the various cell lines selected. Although not 100% conclusive the resultant southern indicates that there is three copies of the VSV-G gene in PI, two of which are probably truncated, and a single complete copy in VSV7.
  • RNA content of the cells was isolated using TRIZOL reagent (GIBCOBRL) and a northern blot was performed using standard methods. Total RNA was isolated from the cells grown at 37°C or at 32° C for four days. This was analysed by northern blotting and 32 P labelled VSV-G DNA probes prepared in the same manner as described for southern blotting ( Figure 24). The amount of signal per lane was quantified using a phosphoimager.
  • RNA content of each lane " was standardised using probes to ribosomal RNAs and then the relative amount of VSV-G messenger RNA was calculated.
  • the ratio of VSV-G mRNA in the cells at 32°C to 37° C was then calculated ( Figure 25).
  • the induction levels were found to be similar in the VSV7 and PI cell lines but a large amount of variation was found between the two gag/pol PI derivatives. The reason for this variation is unknown.
  • RNA samples were isolated from the cells to determine by northern blotting if the increases observed at 32°C were due to effects on translation and/or protein stability or on mRNA levels as was the case for VSV-G.
  • the northern blot was probed as previously described ( Figure 27).
  • Figure 27 two different exposures are shown for each lane, the A lanes were exposed for a shorter time period than the B lanes.
  • Gag/pol mRNA levels were found to increase in a similar manner to the levels of gag/pol protein present.
  • the induction levels were calculated as for the VSV-G mRNA levels, samples being standardised relative to ribosomal RNA levels (Figure 28)
  • the induction levels were of the same order of magnitude as was observed for VSV-G ( Figure 25).
  • VSV7 cell line Similar studies were performed on the VSV7 cell line, however in contrast to the EIAV gag/pol levels, MLV gag/pol levels in this cell line were not found to be higher at 32°C than at 37° C. This difference might give an insight into the mechanism behind the temperature regulation of VSV-G mRNA. Both the VSV-G and EIAV gag/pol genes are transcribed from CMV promoters, whilst the MLV gag/pol is transcribed from an LTR. It is possible that a temperature sensitive mutation in a factor that binds CMV promoters but not MLV LTRs is responsible for the observed phenotype.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne une cellule d'encapsidation et une cellule productrice. La cellule d'encapsidation comprend une première séquence nucléotidique codant une protéine d'enveloppe virale toxique et une deuxième séquence nucléotidique codant une protéine de capside nucléique rétrovirale, l'expression de la première séquence nucléotidique pouvant être régulée dans une plage de température comprise entre 25 et 40 °C. La cellule productrice comprend une cellule d'encapsidation comprenant une séquence virale suffisante pour produire des particules de vecteur viral provenant de la cellule, le titre de particules de vecteur viral obtenu dépendant de l'expression régulée par la température de la protéine d'enveloppe virale toxique.
PCT/GB2000/000766 1999-03-03 2000-03-03 Cellules d'encapsidation pour vecteurs retroviraux WO2000052188A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU29260/00A AU2926000A (en) 1999-03-03 2000-03-03 Packaging cells for retroviral vectors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB9904905.8A GB9904905D0 (en) 1999-03-03 1999-03-03 Cells
GB9904905.8 1999-03-03

Publications (1)

Publication Number Publication Date
WO2000052188A1 true WO2000052188A1 (fr) 2000-09-08

Family

ID=10848893

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2000/000766 WO2000052188A1 (fr) 1999-03-03 2000-03-03 Cellules d'encapsidation pour vecteurs retroviraux

Country Status (3)

Country Link
AU (1) AU2926000A (fr)
GB (1) GB9904905D0 (fr)
WO (1) WO2000052188A1 (fr)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003020931A2 (fr) * 2001-09-01 2003-03-13 Galapagos Genomics N.V. Dosage biologique de choc a interference arn de courte duree et constructions
US6969598B2 (en) 2001-04-30 2005-11-29 Oxford Biomedica (Uk) Limited Methods for producing high titre vectors and compositions used in such methods
US7893036B2 (en) 2001-07-12 2011-02-22 University Of Massachusetts In vivo production of small interfering RNAs that mediate gene silencing
WO2011148194A1 (fr) 2010-05-28 2011-12-01 Oxford Biomedica (Uk) Ltd Administration de vecteurs lentiviraux au cerveau
WO2013061076A1 (fr) 2011-10-28 2013-05-02 Oxford Biomedica (Uk) Limited Construction
WO2016189326A1 (fr) * 2015-05-27 2016-12-01 Oxford Genetics Ltd Lignées cellulaires
EP3192874A1 (fr) 2008-06-18 2017-07-19 Oxford BioMedica (UK) Limited Purification de virus
US9840720B2 (en) 2012-02-13 2017-12-12 Ucl Business Plc Materials and methods relating to packaging cell lines
US9879272B2 (en) 2007-05-25 2018-01-30 22Nd Century Limited, Llc Nucleic acid sequences encoding transcription factors regulating alkaloid biosynthesis and their use in modifying plant metabolism
EP3502260A1 (fr) 2017-12-22 2019-06-26 Oxford BioMedica (UK) Limited Vecteur rétroviral
EP3650548A1 (fr) 2013-12-20 2020-05-13 Oxford BioMedica (UK) Limited Système de production de vecteurs viraux
WO2020183374A1 (fr) 2019-03-10 2020-09-17 Axovant Sciences Gmbh Compositions et procédés de thérapie génique pour le traitement de la maladie de parkinson
WO2021094752A1 (fr) 2019-11-12 2021-05-20 Oxford Biomedica (Uk) Limited Système de production
WO2021160993A1 (fr) 2020-02-13 2021-08-19 Oxford Biomedica (Uk) Limited Production de vecteurs lentiviraux
WO2021181108A1 (fr) 2020-03-13 2021-09-16 Oxford Biomedica (Uk) Limited Vecteurs lentiviraux
WO2021229242A1 (fr) 2020-05-15 2021-11-18 Oxford Biomedica (Uk) Limited Production de vecteur viral
GB202114532D0 (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
WO2022101617A1 (fr) 2020-11-10 2022-05-19 Oxford Biomedica (Uk) Limited Préparation d'une solution de complexes polymère/acide nucléique
WO2023062359A2 (fr) 2021-10-12 2023-04-20 Oxford Biomedica (Uk) Limited Nouveaux éléments régulateurs viraux
WO2023062363A1 (fr) 2021-10-12 2023-04-20 Oxford Biomedica (Uk) Limited Vecteurs lentiviraux
WO2023105235A1 (fr) 2021-12-09 2023-06-15 Oxford Biomedica (Uk) Limited Procédé de purification de vecteurs viraux
WO2024038266A1 (fr) 2022-08-16 2024-02-22 Oxford Biomedica (Uk) Limited Proteines d'enveloppe

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994029440A1 (fr) * 1993-06-04 1994-12-22 The Regents Of The University Of California Production, concentration et transfert efficace de vecteurs retroviraux resultant de la formation de pseudotypes par la proteine g du virus de la stomatite vesiculeuse (vsv)

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994029440A1 (fr) * 1993-06-04 1994-12-22 The Regents Of The University Of California Production, concentration et transfert efficace de vecteurs retroviraux resultant de la formation de pseudotypes par la proteine g du virus de la stomatite vesiculeuse (vsv)

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MACHAMER C. E. ET AL.: "A SINGLE N-LINKED OLIGOSACCHARIDE AT EITHER OF THE TWO NORMAL SITES IS SUFFICIENT FOR TRANSPORT OF VESICULAR STOMATITIS VIRUS G PROTEIN TO THE CELL SURFACE", MOLECULAR AND CELLULAR BIOLOGY, vol. 5, no. 11, 1985, pages 3074 - 3083, XP000929480, ISSN: 0270-7306 *
MACHAMER C. E. ET AL.: "VESICULAR STOMATITIS VIRUS G PROTEINS WITH ALTERED GLYCOSYLATION SITES DISPLAY TEMPERATURE-SENSITIVE INTRACELLULAR TRANSPORT AND ARE SUBJECT TO ABERRANT INTERMOLECULAR DISULFIDE BONDING", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 263, no. 12, 1988, pages 5955 - 5960, XP002143424, ISSN: 0021-9258 *

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6969598B2 (en) 2001-04-30 2005-11-29 Oxford Biomedica (Uk) Limited Methods for producing high titre vectors and compositions used in such methods
US9850487B2 (en) 2001-07-12 2017-12-26 University Of Massachusetts In vivo production of small interfering RNAs that mediate gene silencing
US7893036B2 (en) 2001-07-12 2011-02-22 University Of Massachusetts In vivo production of small interfering RNAs that mediate gene silencing
US10731155B2 (en) 2001-07-12 2020-08-04 University Of Massachusetts In vivo production of small interfering RNAs that mediate gene silencing
US8232260B2 (en) 2001-07-12 2012-07-31 University Of Massachusetts In vivo production of small interfering RNAs that mediate gene silencing
US8530438B2 (en) 2001-07-12 2013-09-10 University Of Massachusetts Vivo production of small interfering RNAs that mediate gene silencing
US8557785B2 (en) 2001-07-12 2013-10-15 University Of Massachusetts In vivo production of small interfering RNAS that mediate gene silencing
US9175287B2 (en) 2001-07-12 2015-11-03 University Of Massachusetts In vivo production of small interfering RNAs that mediate gene silencing
WO2003020931A3 (fr) * 2001-09-01 2003-11-20 Galapagos Genomics Nv Dosage biologique de choc a interference arn de courte duree et constructions
WO2003020931A2 (fr) * 2001-09-01 2003-03-13 Galapagos Genomics N.V. Dosage biologique de choc a interference arn de courte duree et constructions
US9879272B2 (en) 2007-05-25 2018-01-30 22Nd Century Limited, Llc Nucleic acid sequences encoding transcription factors regulating alkaloid biosynthesis and their use in modifying plant metabolism
EP3192874A1 (fr) 2008-06-18 2017-07-19 Oxford BioMedica (UK) Limited Purification de virus
WO2011148194A1 (fr) 2010-05-28 2011-12-01 Oxford Biomedica (Uk) Ltd Administration de vecteurs lentiviraux au cerveau
EP3219801A1 (fr) 2011-10-28 2017-09-20 Oxford BioMedica (UK) Limited Construction
WO2013061076A1 (fr) 2011-10-28 2013-05-02 Oxford Biomedica (Uk) Limited Construction
US9840720B2 (en) 2012-02-13 2017-12-12 Ucl Business Plc Materials and methods relating to packaging cell lines
EP3650548A1 (fr) 2013-12-20 2020-05-13 Oxford BioMedica (UK) Limited Système de production de vecteurs viraux
WO2016189326A1 (fr) * 2015-05-27 2016-12-01 Oxford Genetics Ltd Lignées cellulaires
GB2554316A (en) * 2015-05-27 2018-03-28 Oxford Genetics Ltd Cell Lines
EP3502260A1 (fr) 2017-12-22 2019-06-26 Oxford BioMedica (UK) Limited Vecteur rétroviral
EP3696272A1 (fr) 2017-12-22 2020-08-19 Oxford BioMedica (UK) Limited Vecteur rétroviral
EP3633040A1 (fr) 2017-12-22 2020-04-08 Oxford BioMedica (UK) Limited Vecteur rétroviral
WO2020183374A1 (fr) 2019-03-10 2020-09-17 Axovant Sciences Gmbh Compositions et procédés de thérapie génique pour le traitement de la maladie de parkinson
WO2021094752A1 (fr) 2019-11-12 2021-05-20 Oxford Biomedica (Uk) Limited Système de production
WO2021160993A1 (fr) 2020-02-13 2021-08-19 Oxford Biomedica (Uk) Limited Production de vecteurs lentiviraux
WO2021181108A1 (fr) 2020-03-13 2021-09-16 Oxford Biomedica (Uk) Limited Vecteurs lentiviraux
WO2021229242A1 (fr) 2020-05-15 2021-11-18 Oxford Biomedica (Uk) Limited Production de vecteur viral
WO2022101617A1 (fr) 2020-11-10 2022-05-19 Oxford Biomedica (Uk) Limited Préparation d'une solution de complexes polymère/acide nucléique
EP4335457A2 (fr) 2020-11-10 2024-03-13 Oxford BioMedica (UK) Limited Préparation d'une solution de complexes polymère/acide nucléique
EP4335458A2 (fr) 2020-11-10 2024-03-13 Oxford BioMedica (UK) Limited Préparation d'une solution de complexes polymère/acide nucléique
GB202114530D0 (en) 2021-10-12 2021-11-24 Oxford Biomedica Ltd Retroviral vectors
WO2023062366A1 (fr) 2021-10-12 2023-04-20 Oxford Biomedica (Uk) Limited Vecteurs rétroviraux
WO2023062359A2 (fr) 2021-10-12 2023-04-20 Oxford Biomedica (Uk) Limited Nouveaux éléments régulateurs viraux
WO2023062365A2 (fr) 2021-10-12 2023-04-20 Oxford Biomedica (Uk) Limited Vecteurs lentiviraux
WO2023062363A1 (fr) 2021-10-12 2023-04-20 Oxford Biomedica (Uk) Limited Vecteurs lentiviraux
WO2023062367A1 (fr) 2021-10-12 2023-04-20 Oxford Biomedica (Uk) Limited Vecteurs lentiviraux
GB202114529D0 (en) 2021-10-12 2021-11-24 Oxford Biomedica Ltd Lentiviral vectors
GB202114532D0 (en) 2021-10-12 2021-11-24 Oxford Biomedica Ltd Lentiviral Vectors
WO2023105235A1 (fr) 2021-12-09 2023-06-15 Oxford Biomedica (Uk) Limited Procédé de purification de vecteurs viraux
WO2024038266A1 (fr) 2022-08-16 2024-02-22 Oxford Biomedica (Uk) Limited Proteines d'enveloppe

Also Published As

Publication number Publication date
GB9904905D0 (en) 1999-04-28
AU2926000A (en) 2000-09-21

Similar Documents

Publication Publication Date Title
WO2000052188A1 (fr) Cellules d'encapsidation pour vecteurs retroviraux
JP4979851B2 (ja) 高いタイターで安全な組換えレンチウイルスベクターの作製方法
JP4981231B2 (ja) レトロウイルスパッケージング細胞での発現のためのコドン最適化
JP2020096609A (ja) ウイルスベクター産生系
US7056699B2 (en) Lentiviral LTR-deleted vector
US20170166924A1 (en) Methods for producing and using in vivo pseudotyped retroviruses
Kumar et al. Large-scale production of pseudotyped lentiviral vectors using baculovirus GP64
JP2002508184A (ja) 高力価で安全な組換えレンチウイルスベクター作成の方法及び手段
AU763007B2 (en) Retroviral delivery system
JP4413495B2 (ja) レンチウィルスパッケージング構築物
EP1534847B1 (fr) Vecteur retroviral et lignees cellulaires d'encapsidation stables
ES2627445T3 (es) Partículas de vector de lentivirus resistentes a la inactivación por el complemento
US6969598B2 (en) Methods for producing high titre vectors and compositions used in such methods
CA2849652C (fr) Utilisation de proteines gag non de sous-type b pour l'emballage lentiviral
US6818209B1 (en) Retroviral delivery system
US7135339B2 (en) Methods for producing and using in vivo pseudotyped retroviruses using envelope glycoproteins from lymphocytic choriomeningitis virus (LCMV)
AU773015B2 (en) Lentiviral vectors
US20040033604A1 (en) Recombinant lentiviral vectors pseudotyped in envelopes containing filovirus binding domains
US20220228169A1 (en) Small ruminant lentivirus vector
US20030143205A1 (en) Alphavirus expression systems
WO2001083730A2 (fr) Vecteurs lentiviraux de recombinaison pseudotypes dans des enveloppes contenant des domaines de liaison de filovirus
WO2004044157A2 (fr) Vecteurs pseudotypes avec la gp64 et leurs utilisations
AU2002238584C1 (en) Lentiviral packaging constructs
AU2002246958B2 (en) Vectors and packaging systems for transduction into quiescent cells
JP2001514517A (ja) 修飾泡沫状ウイルスエンベロープタンパク質の発現

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase