WO2011110864A1 - Promoter sequence for dna and viral vectors - Google Patents

Promoter sequence for dna and viral vectors Download PDF

Info

Publication number
WO2011110864A1
WO2011110864A1 PCT/GB2011/050485 GB2011050485W WO2011110864A1 WO 2011110864 A1 WO2011110864 A1 WO 2011110864A1 GB 2011050485 W GB2011050485 W GB 2011050485W WO 2011110864 A1 WO2011110864 A1 WO 2011110864A1
Authority
WO
WIPO (PCT)
Prior art keywords
viral vector
sequence
promoter
nucleic acid
gene
Prior art date
Application number
PCT/GB2011/050485
Other languages
French (fr)
Inventor
David Hamilton Wyllie
Matthew Guy Cottingham
Original Assignee
Isis Innovation 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 Isis Innovation Limited filed Critical Isis Innovation Limited
Publication of WO2011110864A1 publication Critical patent/WO2011110864A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/001Vector systems having a special element relevant for transcription controllable enhancer/promoter combination
    • C12N2830/005Vector systems having a special element relevant for transcription controllable enhancer/promoter combination repressible enhancer/promoter combination, e.g. KRAB
    • C12N2830/006Vector systems having a special element relevant for transcription controllable enhancer/promoter combination repressible enhancer/promoter combination, e.g. KRAB tet repressible
    • 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
    • C12N2830/15Vector systems having a special element relevant for transcription chimeric enhancer/promoter combination
    • 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
    • C12N2830/20Vector systems having a special element relevant for transcription transcription of more than one cistron
    • 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
    • C12N2830/60Vector systems having a special element relevant for transcription from viruses
    • 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
    • C12N2830/80Vector systems having a special element relevant for transcription from vertebrates

Definitions

  • the present invention relates to viral vectors, to methods of making a viral vector, to host cells for use in making viral vectors, and to methods of expressing a gene or genes in a target cell using a single viral vector.
  • Viral vectors are used in multiple applications in both basic biochemical research and medicine. Such vectors may be engineered using recombinant nucleic acid technology and are used to transfer a gene or genes of interest into a target cell, leading to the expression of the gene or genes and the production of the gene product or products encoded. Viral vectors have found application in vaccines against infectious diseases, in the treatment of cancer, and in gene therapy.
  • a promoter sequence is a length of DNA sequence to which RNA polymerase binds in order to begin the process of transcribing the coding sequence of the gene to RNA.
  • CMV human cytomegalovirus
  • SV40 simian virus 40
  • Iva2 adenoviral major late and Iva2 promoters
  • a promoter should be highly active in order to achieve high levels of transcription in the target cell.
  • examples of applications where such expression of multiple genes is desirable or essential include the expression of an adjuvant together with an antigen; the expression of multiple antigens, with one example being the need to express antigens from different pathogen serotypes to develop vaccines with broad coverage; the generation of immune responses against multiple targets; and the expression of a marker gene.
  • the present invention solves one or more of the above technical problems by providing a viral vector comprising a first promoter sequence, wherein the first promoter sequence comprises:
  • xEF1 -alpha SEQ ID NO: 1
  • SEQ ID NO: 1 excludes the presence of any known enhancer elements.
  • the first promoter sequence consists of or comprises a nucleic acid sequence having at least 70% (such as at least 75, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or 100%) nucleic acid sequence identity to the nucleic acid sequence of SEQ ID NO: 1 .
  • the nucleic acid sequence identity with SEQ ID NO: 1 exists over a region of the nucleic acid sequence of SEQ ID NO: 1 that is at least 100 consecutive nucleotides in length (e.g. at least 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, consecutive nucleotides or the entire nucleotide in length).
  • each of said fragments comprises (or consists of) at least 10 consecutive nucleotides (e.g. at least 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 1 10, 1 15, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180 or 185 nucleotides).
  • the at least two fragments of (a) may number at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9 or at least 10 fragments.
  • the overall nucleotide length of the 'at least two fragment aspect' is at least 100 nucleotides, or at least 150 nucleotides, or at least 200 nucleotides, or at least 250 nucleotides, or at least 300 nucleotides, or at least 350 nucleotides in length.
  • a fragment in the context of the 'at least two fragment' aspect (b) of the first promoter sequence, a fragment may be selected from a sequence having least 70% (such as at least 75, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or 100%) nucleic acid sequence identity to any one or more of the following SEQ ID NO: 1 nucleic acid fragments:
  • said fragment comprises (or consists of) at least 21 consecutive nucleotides (e.g. at least 24, 30, 42, 51 , 60, 72, 81 , 90, 102, 1 1 1 , 120, 132, 141 , 150, 162, 171 , 180, 192, 201 , 210, 222, 231 , 240, 252, 261 , 270, 282, 291 , 300, 312, 321 , 330, 342, 351 , 360 or 372 nucleotides) of nucleic acid sequence according to aspect (a).
  • the fragment aspect (c) may include any one or more of the 'at least two' fragments described immediately above for aspect (b).
  • the fragment in the context of the first promoter sequence, has a sequence length that is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of that of the sequence of the full-length nucleic acid sequence SEQ ID NO: 1 .
  • the nucleic acid sequence identity with SEQ ID NO: 1 exists over a region of the nucleic acid sequence of SEQ ID NO: 1 that is at least 20 consecutive nucleotides in length.
  • the region of identity is over a length of SEQ ID NO: 1 that is the same length as the length of the fragment of the invention.
  • the region of identity is over a length of SEQ ID NO: 1 that is at least 125, 150, 175, 200, 225, 250, 275, 300, 325, 350 or 375 consecutive nucleotides of SEQ ID NO: 1.
  • viral vector means any recombinant virus suitable for introducing genetic material into a cell.
  • the first promoter sequence provides a strong promoter, capable of facilitating a high level of transcription.
  • the first promoter sequence is of a size advantageous for use in a viral vector such as an adenoviral vector.
  • the first promoter sequence will not undergo homologous recombination with the human genome.
  • a regulatory consideration when viruses are used in humans, is the potential risk of homologous integration of expression cassettes containing human promoters into the human genome. For this reason, promoters lacking primary sequence homology with human sequences may be preferable for regulatory reasons.
  • the viral vector further comprises an enhancer sequence; wherein the enhancer sequence increases transcription from the first promoter sequence.
  • the term "enhancer sequence” means any nucleic acid sequence, the presence of which increases transcription from the first promoter sequence (for example, by leading to an increase in the number of transcripts produced over a given period of time, in comparison to the number of transcripts produced in the same period of time in the absence of the enhancer).
  • the enhancer sequence may be located anywhere in the viral vector, and may be present in either orientation.
  • the enhancer sequence comprises a nucleic acid sequence having at least 70% identity to the nucleic acid sequence of SEQ ID NO: 2, or a fragment thereof comprising at least 10 consecutive nucleotides thereof.
  • the nucleic acid sequence represented by SEQ ID NO: 2 is based on the enhancer sequence of SV40 virus (Simian Virus 40).
  • the enhancer sequence consists of or comprises a nucleic acid sequence having at least 70% (such as at least 75, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or 100%) nucleic acid sequence identity to the nucleic acid sequence of SEQ ID NO: 2, or a fragment thereof comprising at least 10 consecutive nucleotides thereof.
  • the nucleic acid sequence identity exists over a region of the nucleic acid sequence that is at least 10 consecutive nucleotides in length (e.g. at least 15, 25, 30, 35, 40, 45, 50, or 55 consecutive nucleotides in length).
  • a fragment comprises (or consists of) at least 10 consecutive nucleotides of said nucleic acid sequence (e.g. at least 15, 20, 25, 30, 35, 40, 42, 44, 46, 48, 50, 52 or 54 consecutive nucleotides thereof).
  • a fragment of a nucleic acid sequence has a sequence length that is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of that of the sequence of the full-length nucleic acid sequence.
  • the enhancer sequence comprises or consists of a nucleic acid sequence having at least 70% (such as at least 75, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or 100%) sequence identity to the nucleic acid sequence of SEQ ID NO: 3, or a fragment thereof comprising at least 10 consecutive nucleotides thereof.
  • the nucleic acid sequence represented by SEQ ID NO: 3 is based on the Xenopus laevis EF1 -alpha gene enhancer sequence.
  • the nucleic acid sequence identity exists over a region of the nucleic acid sequence that is at least 10 consecutive nucleotides in length (e.g. at least 15, 25, 30, 35, 40, 45, 50, or 55 consecutive nucleotides in length).
  • a fragment comprises (or consists of) at least 10 consecutive nucleotides of said nucleic acid sequence (e.g. at least 15, 20, 25, 30, 35, 40, 42, 44, 46, 48, 50, 52 or 54 consecutive nucleotides thereof).
  • a fragment of a nucleic acid sequence has a sequence length that is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of that of the sequence of the full-length nucleic acid sequence.
  • the viral vector further comprises at least one gene; wherein the first promoter sequence is operably-linked to the at least one gene, and facilitates the expression thereof.
  • the term "gene” encompasses both protein-coding and non protein-coding genes.
  • the viral vector comprises at least one protein-coding gene.
  • the viral vector comprises at least one non protein-coding gene.
  • the non protein-coding gene may encode an RNA.
  • the non protein-coding gene encodes a small interfering RNA (siRNA).
  • siRNA small interfering RNA
  • an antigen suitable for use in the present invention is the METRAP antigen.
  • the gene encodes the METRAP antigen.
  • An example of an adjuvant suitable for use in the present invention is granulocyte-macrophage colony-stimulating factor (GM-CSF).
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • tumour-specific antigen is a protein or molecule which is either unique to a tumour cell or is present in the tumour cell in much greater abundance compared to a non- tumour cell. It is evident from the above that any gene can be expressed using the viral vector of the present invention, and that there is no intention to limit the present invention to viral vectors expressing any one particular gene. A skilled person will be familiar with other enzyme, reporter and receptor genes which would be suitable for use in the present invention.
  • operably-linked means that the nucleic acid sequences being linked are arranged so that they function in concert for their intended purposes - for example, transcription initiates in the promoter and proceeds through the coding polynucleotide segment to the terminator.
  • transcription may be initiated in the first promoter sequence and proceed through the coding sequence of the at least one gene, leading to the expression of said gene.
  • the viral vector further comprises a second promoter sequence; wherein the first promoter sequence and the second promoter sequence are different.
  • the second promoter sequence is any promoter sequence that is not the above- defined first promoter sequence.
  • the second promoter sequence is a promoter sequence lacking sequence homology with the first promoter sequence, to the extent that homologous recombination between the first and second promoter sequences cannot take place.
  • the first promoter sequence is operably-linked to a first gene, and facilitates the expression thereof; and the second promoter sequence is operably-linked to a second gene, and facilitates the expression thereof.
  • the viral vector is a bicistronic viral vector.
  • the viral vector comprises at least one additional promoter sequence operably-linked to an additional gene, wherein the additional promoter sequence facilitates the expression of the additional gene.
  • the viral vector may comprise a third promoter sequence, a fourth promoter sequence, or a fifth promoter sequence, or additional promoter sequences beyond a fifth promoter sequence, each additional promoter sequence operably-linked to a gene and facilitating the expression thereof.
  • the viral vector is a multicistronic vector.
  • the second promoter sequence is selected from the group consisting of: human CMV promoter, simian CMV promoter, murine or other mammalian CMV promoters, promoters derived from other herpes viruses, promoters from other types of virus, and mammalian promoters.
  • the viral vector is any virus which can serve as a viral vector. Suitable viruses are those which infect the species of interest, can be propagated in vitro, and can be modified by recombinant nucleotide technology known in the art.
  • the viral vector is selected from the group consisting of: an adenovirus vector; an adeno-associated virus vector; a pox virus vector, such as a fowlpox virus vector; an alpha virus vector; a bacloviral vector; a herpes virus vector; a retrovirus vector, such as a lentivirus vector; a Modified Vaccinia virus Ankara vector; a Ross River virus vector; a Sindbis virus vector; a Semliki Forest virus vector; and a Venezuelan Equine Encephalitis virus vector.
  • the viral vector is an adenovirus vector.
  • the viral vector is an adenovirus vector comprising a first promoter sequence, the first promoter sequence being as defined above.
  • the viral vector further comprises at least one repressor binding site sequence.
  • the repressor binding site sequence may be introduced into the viral vector by recombinant DNA technology.
  • the repressor binding site sequence may be associated with the first promoter sequence such that it is able to control the expression of a gene operably-linked to the first promoter sequence.
  • binding of a repressor protein to the repressor binding site enables negative regulation of the first promoter sequence, decreasing or suppressing expression of a gene operably-linked to said promoter.
  • the repressor protein is produced by the host cell in which the viral vector of the present invention is grown.
  • An example of a suitable repressor binding site sequence is the tetracycline operator site sequence. This sequence is able to bind tetracycline repressor proteins, so decreasing gene expression from an associated promoter sequence.
  • the viral vector includes an artificial repressible promoter sequence.
  • suitable repressor binding site sequences are: a Lac operator, a Mutant Lac operator, and a LexA operator.
  • the viral vector further comprises an additional repressor binding site sequence for each additional promoter sequence beyond the first promoter sequence.
  • the at least one repressor binding site sequence comprises a nucleic acid sequence having at least 70% sequence identity to the nucleic acid sequence of SEQ ID NO: 4, or a fragment thereof comprising at least 10 consecutive nucleotides thereof.
  • the nucleic acid sequence represented by SEQ ID NO: 4 is based on the tetracycline operator site.
  • the viral vector further comprises two tetracycline operator sites.
  • the at least one repressor binding site sequence comprises or consists of a nucleic acid sequence having at least 70% (such as at least 75, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or 100%) nucleic acid sequence identity to the nucleic acid sequence of SEQ ID NO: 4, or a fragment thereof comprising at least 10 consecutive nucleotides thereof.
  • the nucleic acid sequence identity exists over a region of the nucleic acid sequence that is at least 10 consecutive nucleotides in length (e.g. at least 15, 25, 30, 35, 40, 45, 50, or 55 consecutive nucleotides in length).
  • a fragment comprises (or consists of) at least 10 consecutive nucleotides of said nucleic acid sequence (e.g. at least 15, 20, 25, 30, 35, 40, 42, 44, 46, 48, 50, 52 or 54 consecutive nucleotides thereof).
  • a fragment of a nucleic acid sequence has a sequence length that is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of that of the sequence of the full-length nucleic acid sequence.
  • the viral vector comprises at least one repressor binding site sequence (for example a tetracycline operator site) inserted downstream of the TATA box of the first promoter sequence.
  • the number of repressor binding sites may be two, or a number greater than two.
  • the viral vector comprises two repressor binding sites (e.g. the tetracycline operator) inserted downstream of the TATA box of the first promoter sequence.
  • a tetracycline operator site is able to bind the tetracycline repressor protein; the binding of this protein to the operator site acts to inhibit transcription from the proximal promoter.
  • the tetracycline repressor protein is no longer bound to the operator sequence and transcription from the proximal promoter sequence is no longer repressed.
  • the viral vector comprises at least one repressor binding site sequence (for example a tetracycline operator site) inserted downstream of the TATA box of the second promoter sequence.
  • the number of repressor binding sites may be two, or a number greater than two.
  • the viral vector comprises two repressor binding sites (e.g. the tetracycline operator) inserted downstream of the TATA box of the second promoter sequence.
  • the invention provides a method of making a viral vector, comprising providing a nucleic acid, wherein the nucleic acid comprises a nucleic acid sequence encoding a viral vector (as described above); transfecting a host cell with the nucleic acid; culturing the host cell under conditions suitable for the expression of the nucleic acid; and obtaining the viral vector from the host cell.
  • the nucleic acid comprising a sequence encoding a viral vector may be generated by the use of any technique for manipulating and generating recombinant nucleic acid known in the art.
  • "transfecting” may mean any non-viral method of introducing nucleic acid into a cell.
  • the nucleic acid may be any nucleic acid suitable for transfecting a host cell.
  • the nucleic acid is a plasmid.
  • the host cell may be any cell in which a viral vector (as described above) may be grown.
  • culturing the host cell under conditions suitable for the expression of the nucleic acid means using any cell culture conditions and techniques known in the art which are suitable for the chosen host cell, and which enable the viral vector to be produced in the host cell.
  • obtaining the viral vector means using any technique known in the art that is suitable for separating the viral vector from the host cell.
  • the host cells are lysed to release the viral vector.
  • the viral vector may subsequently be isolated and purified using any suitable method or methods known in the art.
  • the invention provides a host cell, comprising a viral vector (as described above).
  • the host cell may be any cell in which a viral vector (as described above) may be grown.
  • the host cell is selected from the group consisting of: a 293 cell, a CHO cell, a CCL81.1 cell, a Vera cell, a HELA cell, a Per.C6 cell, and a BHK cell.
  • the invention provides a method of expressing in a target cell at least one protein, comprising providing a viral vector (as described above); and introducing the viral vector into a target cell.
  • the method of expressing in a target cell at least one protein may be carried out on a target cell in vitro, in vivo or ex vivo.
  • the target cell may be part of an in vitro cell culture, or part of a subject in vivo, or part of an ex vivo organ or tissue.
  • the invention provides an in vitro method of expressing in a target cell at least one protein, comprising providing a viral vector (as described above); and introducing the viral vector into a target cell.
  • the viral vector (as described above) is provided according to the method of making a viral vector described above.
  • the viral vector may be introduced into the target cell according to any suitable method known in the art.
  • the viral vector of the present invention has multiple utilities, which are described below.
  • the invention provides a viral vector (as described above) for use in medicine.
  • the invention provides a viral vector (as described above), for use in gene therapy.
  • a viral vector as described above
  • Methods and applications of gene therapy are known in the art.
  • the viral vector is for use in any method which aims to prevent, treat or cure a disease or diseases through the introduction of novel genetic material into the cells of a subject.
  • the invention provides a viral vector for use in the treatment of cancer.
  • the viral vector (as described above), for use in the treatment of cancer may comprise at least one gene encoding a tumour-specific antigen (such tumour-specific antigen being as described above).
  • the tumour- specific antigen may be displayed on the external surface of the tumour cell, or may be present internally.
  • the viral vector (as described above), for use in the treatment of cancer is used wherein the treatment comprises administering the viral vector to a subject and stimulating in the subject an immunogenic response against a tumour cell or cells present in the subject.
  • the subject's immune system may be stimulated to attack a tumour present in the subject.
  • the viral vector for use in the treatment of cancer, may comprise at least one gene encoding a gene product which has anti-tumour or anti-cancer properties.
  • the gene product may be a nucleic acid (for example, a small interfering RNA) or a protein.
  • the gene product may inhibit the proliferation and/or division of tumour/cancer cells.
  • the gene product may cause the death of tumour/cancer cells (for example, via necrosis or apoptosis).
  • the invention provides a viral vector (as described above), for use in the treatment of allergy.
  • the viral vector (as described above), for use in the treatment of allergy may further comprise a gene encoding an allergen.
  • the viral vector (as described above), for use in the treatment of allergy may be used wherein the treatment of allergy comprises a prophylactic treatment to prevent the development of an allergy in a subject, or a therapeutic treatment to reduce or abolish the symptoms of an allergy in a subject.
  • the invention provides a viral vector (as described above), for use in stimulating or inducing an immune response in a subject.
  • stimulating or inducing an immune response in a subject comprises administering to the subject a viral vector (as described above).
  • the viral vector (as described above), for use in stimulating or inducing an immune response in a subject comprises at least one gene encoding an antigen.
  • the antigen is the METRAP antigen.
  • the viral vector (as described above), for use in stimulating or inducing an immune response in a subject comprises at least one gene encoding an adjuvant.
  • the viral vector (as described above), for use in stimulating or inducing an immune response in a subject comprises a first gene encoding an antigen, and a second gene encoding an adjuvant.
  • stimulating or inducing an immune response in a subject comprises administering a viral vector (as described above) to a subject, wherein said viral vector is administered substantially prior to, simultaneously with or subsequent to another immunogenic composition.
  • the invention provides a viral vector (as described above), for use in the treatment or prevention of at least one infectious disease.
  • the at least one infectious disease is selected from the group consisting of diseases caused by: Plasmodia, influenza viruses, Mycobacterium tuberculosis, Mycobacterium bovis, other Mycobacteria, hepatitis C virus, other flaviviruses, hepatitis B virus, human immunodeficiency virus, other retroviruses, Staphylococcus aureus, other Staphylococci, Streptococcus pneumoniae, Streptococcus pyogenes, other Streptococci, Haemophilus influenzae, Neisseria meningitides.
  • the treatment or prevention of at least one infectious disease comprises administering to a subject a viral vector (as described above) wherein said viral vector is administered substantially prior to, simultaneously with or subsequent to another immunogenic composition.
  • a viral vector as described above
  • Prior, simultaneous and sequential administration regimes are discussed in more detail below.
  • the viral vector of the present invention may be useful for inducing a range of immune responses and may therefore be useful in methods for treating a range of diseases.
  • treatment or “treating” embraces therapeutic or preventative/prophylactic measures, and includes post-infection therapy and amelioration of an infectious disease.
  • preventing includes preventing the initiation of an infectious disease and/or reducing the severity or intensity of an infectious disease.
  • a viral vector of the invention may be administered to a subject (typically a mammalian subject such as a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) already having an infectious disease, to treat or prevent said infectious disease.
  • a subject typically a mammalian subject such as a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject
  • the subject is suspected of having come into contact with an infectious disease (or the disease-causing agent), or has had known contact with an infectious disease (or the disease-causing agent), but is not yet showing symptoms of exposure to said infectious disease (or said disease-causing agent).
  • a viral vector of the invention can cure, delay, reduce the severity of, or ameliorate one or more symptoms of, the infectious disease; and/or prolong the survival of a subject beyond that expected in the absence of such treatment.
  • a viral vector of the invention may be administered to a subject (e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) who may ultimately contract an infectious disease, in order to prevent, cure, delay, reduce the severity of, or ameliorate one or more symptoms of, said infectious disease; or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.
  • a subject has previously been exposed to an infectious disease.
  • the subject may have had an infectious disease in the past (but is optionally not currently infected with the disease-causing agent of the infectious disease).
  • the subject may be latently infected with an infectious disease.
  • the subject may have been vaccinated against said infectious disease in the past.
  • the treatments and preventative therapies in which viral vectors of the present invention may be used are applicable to a variety of different subjects of different ages.
  • the therapies are applicable to children (e.g. infants, children under 5 years old, older children or teenagers) and adults.
  • the therapies are applicable to immature subjects (e.g. calves, piglets, foals) and mature/adult subjects.
  • the treatments and preventative therapies of the present invention are applicable to subjects who are immunocompromised or immunosuppressed (e.g. human patients who have HIV or AIDS, or other animal patients with comparable immunodeficiency diseases), subjects who have undergone an organ transplant, bone marrow transplant, or who have genetic immunodeficiencies.
  • a "vaccine” is a formulation that, when administered to an animal subject such as a mammal (e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) stimulates a protective immune response against an infectious disease.
  • the immune response may be a humoral and/or a cell-mediated immune response.
  • the vaccine may stimulate B- cells and/or T-cells.
  • a vaccine of the invention can be used, for example, to protect an animal from the effects of an infectious disease (for example, malaria, influenza or tuberculosis).
  • vaccine is herein used interchangeably with the terms “therapeutic/prophylactic composition”, “formulation”, “antigenic composition”, or “medicament”.
  • the invention provides a vaccine composition, comprising a viral vector (as described above); and a pharmaceutically acceptable carrier.
  • the vaccine of the invention in addition to a pharmaceutically acceptable carrier can further be combined with one or more of a salt, excipient, diluent, adjuvant, immunoregulatory agent and/or antimicrobial compound.
  • the invention provides an immunological composition, comprising a viral vector (as described above); and a pharmaceutically acceptable carrier.
  • the immunological composition in addition to a pharmaceutically acceptable carrier can further be combined with one or more of a salt, excipient, diluent, adjuvant, immunoregulatory agent and/or antimicrobial compound.
  • the invention provides a pharmaceutical composition, comprising a viral vector (as described above); and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition in addition to a pharmaceutically acceptable carrier can further be combined with one or more of a salt, excipient, diluent, adjuvant, immunoregulatory agent and/or antimicrobial compound.
  • the viral vector may be formulated into a vaccine, immunogenic composition or pharmaceutical composition as neutral or salt forms.
  • Pharmaceutically acceptable salts include acid addition salts formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or with organic acids such as acetic, oxalic, tartaric, maleic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • immunogenic compositions are generally by conventional routes e.g. intravenous, subcutaneous, intraperitoneal, or mucosal routes.
  • the administration may be by parenteral administration; for example, a subcutaneous or intramuscular injection.
  • immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention may be prepared as injectables, either as liquid solutions or suspensions.
  • Solid forms suitable for solution in, or suspension in, liquid prior to injection may alternatively be prepared.
  • the preparation may also be emulsified, or the peptide encapsulated in liposomes or microcapsules.
  • the active ingredients are often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like and combinations thereof.
  • excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like and combinations thereof.
  • the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, and/or pH buffering agents.
  • Vaccine and immunological compositions of the present invention may comprise adjuvants which enhance the effectiveness of the vaccine or immunological composition.
  • the carrier is a pharmaceutically-acceptable carrier.
  • Non-limiting examples of pharmaceutically acceptable carriers include water, saline, and phosphate-buffered saline.
  • the composition is in lyophilized form, in which case it may include a stabilizer, such as bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • additional adjuvants which may be effective include but are not limited to: complete Freunds adjuvant (CFA), Incomplete Freunds Adjuvant (IVA), Saponin, a purified extract fraction of Saponin such as Quil A, a derivative of Saponin such as QS-21 , lipid particles based on Saponin such as ISCOM/ISCOMATIX, E.
  • CFA complete Freunds adjuvant
  • IVA Incomplete Freunds Adjuvant
  • Saponin a purified extract fraction of Saponin such as Quil A
  • QS-21 a derivative of Saponin
  • lipid particles based on Saponin such as ISCOM/ISCOMATIX
  • coli heat labile toxin (LT) mutants such as LTK63 and/or LTK72, aluminium hydroxide, N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr- MDP), N-acetyl-nor-muramyl-L-alanyl-D-isoglutamine (CGP 1 1637, referred to as nor-MDP), N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1 '-2'- dipalmitoyl-sn-glycero-3-hydroxyphosphoryl oxy)-ethylamine (CGP 19835A, referred to as MTP-PE), and RIBI, which contains three components extracted from bacteria, monophosphoryl lipid A, trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2 % squalene/Tween
  • buffering agents include, but are not limited to, sodium succinate (pH 6.5), and phosphate buffered saline (PBS; pH 6.5 and 7.5).
  • Additional formulations which are suitable for other modes of administration include suppositories and, in some cases, oral formulations or formulations suitable for distribution as aerosols.
  • suppositories traditional binders and carriers may include, for example, polyalkylene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in the range of 0.5% to 10%, preferably 1 %-2%.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders.
  • the viral vectors of the present invention may be desired to direct the viral vectors of the present invention (as described above) to the respiratory system of a subject; for example, for use in the treatment or prevention of a respiratory disease, or for the targeting of a gene therapy to the respiratory system (such as to the lungs).
  • Efficient transmission of a therapeutic/prophylactic composition or medicament to the site of infection in the lungs may be achieved by oral or intra-nasal administration.
  • Formulations for intranasal administration may be in the form of nasal droplets or a nasal spray.
  • An intranasal formulation may comprise droplets having approximate diameters in the range of 100-5000 ⁇ , such as 500-4000 ⁇ , 1000-3000 ⁇ or 100-1000 ⁇ .
  • the droplets may be in the range of about 0.001 -100 ⁇ , such as 0.1 -50 ⁇ or 1 .0-25 ⁇ , or such as 0.001 -1 ⁇ .
  • the therapeutic/prophylactic formulation or medicament may be an aerosol formulation.
  • the aerosol formulation may take the form of a powder, suspension or solution.
  • the size of aerosol particles is relevant to the delivery capability of an aerosol. Smaller particles may travel further down the respiratory airway towards the alveoli than would larger particles.
  • the aerosol particles have a diameter distribution to facilitate delivery along the entire length of the bronchi, bronchioles, and alveoli.
  • the particle size distribution may be selected to target a particular section of the respiratory airway, for example the alveoli.
  • the particles may have diameters in the approximate range of 0.1 - 50 ⁇ , preferably 1 -25 ⁇ , more preferably 1 -5 ⁇ .
  • Aerosol particles may be for delivery using a nebulizer (e.g. via the mouth) or nasal spray.
  • An aerosol formulation may optionally contain a propellant and/or surfactant.
  • Intra-nasal vaccination engages both T- and B-cell mediated effector mechanisms in nasal and bronchus associated mucosal tissues, which differ from other mucosa-associated lymphoid tissues.
  • the protective mechanisms invoked by the intranasal route of administration may include: the activation of T- lymphocytes with preferential lung homing; up-regulation of co-stimulatory molecules (e.g. B7.2); and/or activation of macrophages or secretory IgA antibodies.
  • Intranasal delivery of viral vectors of the invention may facilitate the invoking of a mucosal antibody response, which is favoured by a shift in the T-cell response toward the Th2 phenotype which helps antibody production.
  • a mucosal response is characterised by enhanced IgA production
  • a Th2 response is characterised by enhanced IL-4 production.
  • the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention comprise a pharmaceutically acceptable carrier, and optionally one or more of a salt, excipient, diluent and/ or adjuvant.
  • the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention may comprise one or more immunoregulatory agents selected from, for example, immunoglobulins, antibiotics, interleukins (e.g. IL-2, IL-12), and/or cytokines (e.g. IFNy).
  • the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention may comprise one or more antimicrobial compounds, (for example, conventional anti-tuberculosis drugs such as rifampicin, isoniazid, ethambutol or pyrizinamide).
  • the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention may be given in a single dose schedule (i.e. the full dose is given at substantially one time).
  • the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention may be given in a multiple dose schedule.
  • a multiple dose schedule is one in which a primary course of treatment (e.g. vaccination) may be with 1 -6 separate doses, followed by other doses given at subsequent time intervals required to maintain and or reinforce the immune response, for example (for human subjects), at 1 -4 months for a second dose, and if needed, a subsequent dose(s) after a further 1 -4 months.
  • the dosage regimen will be determined, at least in part, by the need of the individual and be dependent upon the judgment of the practitioner (e.g. doctor or veterinarian).
  • Simultaneous administration means administration at (substantially) the same time.
  • Sequential administration of two or more compositions/therapeutic agents/vaccines means that the compositions/therapeutic agents/vaccines are administered at (substantially) different times, one after the other.
  • the vaccine of the present invention may be administered as part of a 'prime-boost' vaccination regime.
  • the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention can be administered to a subject such as a mammal (e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) in conjunction with (simultaneously or sequentially) one or more immunoregulatory agents selected from, for example, immunoglobulins, antibiotics, interleukins (e.g. IL-2, IL-12), and/or cytokines (e.g. IFNy).
  • a mammal e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject
  • immunoregulatory agents selected from, for example, immunoglobulins, antibiotics, interleukins (e.g. IL-2, IL-12), and/or cytokines (e.g. IFN
  • the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention can be administered to a subject such as a mammal (e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) in conjunction with (simultaneously or sequentially) one or more antimicrobial compounds, such as conventional anti-tuberculosis drugs (e.g. rifampicin, isoniazid, ethambutol or pyrizinamide).
  • a mammal e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject
  • one or more antimicrobial compounds such as conventional anti-tuberculosis drugs (e.g. rifampicin, isoniazid, ethambutol or pyrizinamide).
  • the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations may contain 5% to 95% of active ingredient, such as at least 10% or 25% of active ingredient, or at least 40% of active ingredient or at least 50, 55, 60, 70 or 75% active ingredient.
  • the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations are administered in a manner compatible with the dosage formulation, and in such amount as will be prophylactically and/or therapeutically effective.
  • an "effective amount” is a dosage or amount that is sufficient to achieve a desired biological outcome.
  • a “therapeutically effective amount” is an amount which is effective, upon single or multiple dose administration to a subject (such as a mammal - e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) for treating, preventing, curing, delaying, reducing the severity of, ameliorating at least one symptom of a disorder or recurring disorder, or prolonging the survival of the subject beyond that expected in the absence of such treatment.
  • the quantity of active ingredient to be administered depends on the subject to be treated, capacity of the subject's immune system to generate a protective immune response, and the degree of protection desired. Precise amounts of active ingredient required to be administered may depend on the judgment of the practitioner and may be particular to each subject.
  • nucleic acid sequence and “polynucleotide” are used interchangeably and do not imply any length restriction.
  • the terms “nucleic acid” and “nucleotide” are used interchangeably.
  • the terms “nucleic acid sequence” and “polynucleotide” embrace DNA (including cDNA) and RNA sequences.
  • the polynucleotide sequences of the present invention include nucleic acid sequences that have been removed from their naturally occurring environment, recombinant or cloned DNA isolates, and chemically synthesized analogues or analogues biologically synthesized by heterologous systems.
  • the polynucleotides of the present invention may be prepared by any means known in the art. For example, large amounts of the polynucleotides may be produced by replication in a suitable host cell.
  • the natural or synthetic DNA fragments coding for a desired fragment will be incorporated into recombinant nucleic acid constructs, typically DNA constructs, capable of introduction into and replication in a prokaryotic or eukaryotic cell.
  • DNA constructs will be suitable for autonomous replication in a unicellular host, such as yeast or bacteria, but may also be intended for introduction to and integration within the genome of a cultured insect, mammalian, plant or other eukaryotic cell lines.
  • the polynucleotides of the present invention may also be produced by chemical synthesis, e.g. by the phosphoramidite method or the triester method, and may be performed on commercial automated oligonucleotide synthesizers.
  • a double- stranded fragment may be obtained from the single stranded product of chemical synthesis either by synthesizing the complementary strand and annealing the strand together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence.
  • the term "isolated" in the context of the present invention denotes that the polynucleotide sequence has been removed from its natural genetic milieu and is thus free of other extraneous or unwanted coding sequences (but may include naturally occurring 5' and 3' untranslated regions such as promoters and terminators), and is in a form suitable for use within genetically engineered protein production systems. Such isolated molecules are those that are separated from their natural environment. In view of the degeneracy of the genetic code, considerable sequence variation is possible among the polynucleotides of the present invention. Degenerate codons encompassing all possible codons for a given amino acid are set forth below: Amino Acid Codons Degenerate Codon
  • degenerate codon representative of all possible codons encoding each amino acid.
  • some polynucleotides encompassed by the degenerate sequence may encode variant amino acid sequences, but one of ordinary skill in the art can easily identify such variant sequences by reference to the amino acid sequences of the present invention.
  • a “variant" nucleic acid sequence has substantial homology or substantial similarity to a reference nucleic acid sequence (or a fragment thereof).
  • a nucleic acid sequence or fragment thereof is “substantially homologous" (or “substantially identical") to a reference sequence if, when optimally aligned (with appropriate nucleotide insertions or deletions) with the other nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 70%, 75%, 80%, 82, 84, 86, 88, 90, 92, 94, 96, 98 or 99% of the nucleotide bases. Methods for homology determination of nucleic acid sequences are known in the art.
  • a "variant" nucleic acid sequence is substantially homologous with (or substantially identical to) a reference sequence (or a fragment thereof) if the "variant" and the reference sequence they are capable of hybridizing under stringent (e.g. highly stringent) hybridization conditions.
  • Nucleic acid sequence hybridization will be affected by such conditions as salt concentration (e.g. NaCI), temperature, or organic solvents, in addition to the base composition, length of the complementary strands, and the number of nucleotide base mismatches between the hybridizing nucleic acids, as will be readily appreciated by those skilled in the art.
  • Stringent temperature conditions are preferably employed, and generally include temperatures in excess of 30°C, typically in excess of 37°C and preferably in excess of 45°C.
  • Stringent salt conditions will ordinarily be less than 1000 mM, typically less than 500 mM, and preferably less than 200 mM.
  • the pH is typically between 7.0 and 8.3. The combination of parameters is much more important than any single parameter.
  • preferential codon usage refers to codons that are most frequently used in cells of a certain species, thus favouring one or a few representatives of the possible codons encoding each amino acid.
  • the amino acid threonine (Thr) may be encoded by ACA, ACC, ACG, or ACT, but in mammalian host cells ACC is the most commonly used codon; in other species, different Thr codons may be preferential.
  • Preferential codons for a particular host cell species can be introduced into the polynucleotides of the present invention by a variety of methods known in the art. Introduction of preferential codon sequences into recombinant DNA can, for example, enhance production of the protein by making protein translation more efficient within a particular cell type or species.
  • the nucleic acid sequence is codon optimized for expression in a host cell.
  • a "fragment" of a polynucleotide of interest comprises a series of consecutive nucleotides from the sequence of said full-length polynucleotide.
  • a "fragment" of a polynucleotide of interest may comprise (or consist of) at least 30 consecutive nucleotides from the sequence of said polynucleotide (e.g. at least 35, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800 850, 900, 950 or 1000 consecutive nucleic acid residues of said polynucleotide).
  • a fragment may include at least one antigenic determinant and/or may encode at least one antigenic epitope of the corresponding polypeptide of interest.
  • SEQ ID NO: 1 Core EF1 -alpha promoter sequence from Xenopus laevis.
  • SEQ ID NO: 2 Enhancer sequence from SV40.
  • SEQ ID NO: 3 Enhancer sequence from EF1 -alpha gene of Xenopus laevis.
  • SEQ ID NO: 4 Tetracycline operator site sequence.
  • SEQ ID NO: 5 Full length sequence of Xenopus laevis EF1 -alpha promoter.
  • SEQ ID NO: 6 Core Xenopus laevis EF1 -alpha promoter with tetracycline operator sites.
  • SEQ ID NO 7 Human CMV promoter containing intron A.
  • SEQ ID NO 1 1 Enhanced green fluorescent protein (EGFP).
  • SEQ ID NO 16 Synthetic intron from pCI (Promega).
  • SEQ ID NO 17 Rabbit beta-globin intron.
  • SEQ ID NO 18 Human EF1 -alpha promoter.
  • SEQ ID NO 19 Human EF1 -alpha intron.
  • SEQ ID NO: 2 Enhancer sequence from SV40.
  • SEQ ID NO: 3 Enhancer sequence from EF1 -alpha gene of Xenopus laevis.
  • SEQ ID NO: 4 Tetracycline operator site sequence.
  • SEQ ID NO: 6 Core Xenopus laevis EF1 -alpha promoter with tetracycline operator sites.
  • SEQ ID NO: 7 Human CMV promoter containing intron A.
  • SEQ ID NO: 8 Simian CMV IE94 gene promoter.
  • SEQ ID NO: 9 Human CMV immediate-early promoter.
  • SEQ ID NO: 1 Enhanced green fluorescent protein (EGFP).
  • SEQ ID NO: 12 Bovine growth hormone polyadenylation signal.
  • AAACTTGTTC TATAGTGTCA CCTAAATAGG CCTAATGGTC ATAGCTGTTT CCTGTGTGAA 900
  • ATGTACTGCC AAAACCGCAT CACCATGGTA ATAGCGATGA CTAATACGTA GATGTACTGC 1260
  • GATACCGCGA GATACACGCT CACCGGCTCC AGATTTATCA GCAATAAACC AGCCAGCCGG 2940
  • SEQ ID NO: 16 Synthetic intron from pCI (Promega).
  • SEQ ID NO: 17 Rabbit beta-globin intron.
  • SEQ ID NO: 18 Human EF1 -alpha promoter.
  • SEQ ID NO: 19 Human EF1 -alpha intron.
  • a series of vectors were constructed expressing Photinus luciferase under the control of a series of promoters.
  • Some promoters contained natural or synthetic splice signals (Table 1 ). These included the human CMV immediate early cassette 94 promoter (hCMV), the human EF1 a promoter (hEF1 ), the simian CMV immediate early 94 promoter (sCMV), and the Xenopus EF1 a promoter(Johnson and Krieg 1994; Johnson and Krieg 1995).
  • Some constructs also contained the SV40 enhancer.
  • a schematic organisation of the various promoters tested is shown above. Numbers before each construct are plasmid identifiers; sequences of the promoter and polyadenylation signal in each vector are shown in table 1.
  • Immunofluorescent detection of the products of both gene cassettes was performed following infection of 293 cells (in which the virus replicates) with limiting quantities of virus. Expression of both proteins was observed in all cells examined; representative cells are shown.
  • AdC63-METRAP-xEF1 -GFP 2 weeks after immunisation with AdC63-METRAP-xEF1 -GFP, AdC63-METRAP- xEF1 -GFP-SV40, or AdC63-METRAP, spleen ELISPOT assays were performed using peptides present in ME-TRAP or EGFP.
  • the examples are in three parts:
  • a series of reporter vectors were constructed, expressing Photinus luciferase under the control of a series of promoters.
  • Xenopus EF1 a promoter variants were constructed by a combination of gene synthesis and conventional subcloning.
  • the IE94 gene promoter from simian CMV which is a strong promoter of similar function to the widely used human immediate early cytomegalovirus promoter. Sequence details are in Table 1 . These were transiently transfected into HeLa cells (ATCC CCL-2) or NIH-3T3 cells (ATCC CRL-1658), using an internal control HSV-TK expression plasmid (Promega, pGL4.74), using Superfect (Qiagen). 16-24 hours after transfection, reporter activity was assayed using a Dual Luciferase kit (Promega). Results were expressed as luciferase activities relative to that produced by pGL4.74.
  • Bicistronic adenoviruses were constructed.
  • METRAP was converted into a bicistronic plasmid expressing METRAP by addition of a cassette expressing the enhanced green fluorescent protein (EGFP, Clontech).
  • the vector is illustrated schematically below ( Figure 2).
  • a shuttle plasmid (pKan) was constructed for this purpose.
  • METRAP plasmid was linearised at the unique AsiSI restriction site between the Bovine Growth hormone polyadenylation signal and the adenoviral gene IX, and dephosphorylated.
  • Adenoviral plasmids were linearised, and 5mcg of DNA was transfected into 293 cells (ATCC CRL-1573) using Lipofectamine 2000 (Invitrogen), according to the manufacturer's recommendation.
  • Cell growth media consisted of Dulbecco's Modified Eagle Medium (DMEM), supplemented by penicillin, streptomycin, 2mM L-glutamine and 10% fetal bovine serum (all form Life Technologies).
  • DMEM Dulbecco's Modified Eagle Medium
  • Viruses were grown for five serial passages in 293 cells, and purified by Caesium chloride centrifugation after a varying number of passages. Quantitative PCR was performed genome copy number, and infectivity determined using anti- hexon immunostaining.
  • 293 cells were infected with AdC63 expressing METRAP, or METRAP together with EGFP under the control of the sCMV promoter, xEF1 promoter, or xEF1 promoter-SV40 enhancer at an MOI of 0.1 . 24 hours later, cells were fixed with 2% paraformaldehyde in 0.1 M Tris-HCI pH 8.0, and permeabilised with 0.1 M Tris-HCI pH 8.0, 0.05% Tween 20. Immunostaining was performed with a mouse anti-METRAP antiserum (a kind gift of Okairos AG, Italy) at 1 :200, and rabbit anti-mouse Alexa 647 (Invitrogen) was used as a second layer. Immunofluorescence images were obtained using an Eclipse microscope (Nikon), standard filters and a CCD camera (Hanamatsu).
  • mice Female Balb/c mice (Harlan UK Limited) were used in all experiments under the terms of the U.K. Home Office Animals (Scientific Procedures) Act Project Licence. Animals, 6-1 1 week of age, were immunised with 2.5 ⁇ 10 ⁇ 7 ihu i.d., and ⁇ -IFN ELISPOT assays performed on splenocytes 14 days later.
  • a single cell suspension of splenocytes was generated mechanically, and erythrocytes lysed by resuspension of ACK buffer (0.15M NH 4 CI, 1 mM KHC0 3 , 0.1 M Na 2 EDTA, pH 7.3), followed by resuspension in growth media (a-MEM, 10% FCS, 2mM L- glutamine, 0.1 % ⁇ -mercaptoethanol, 100U penicillin and 100ug streptomycin) (all from Life Technologies).
  • ACK buffer 0.15M NH 4 CI, 1 mM KHC0 3 , 0.1 M Na 2 EDTA, pH 7.3
  • growth media a-MEM, 10% FCS, 2mM L- glutamine, 0.1 % ⁇ -mercaptoethanol, 100U penicillin and 100ug streptomycin
  • Cells were plated on Multiscreen® filter plates (Milipore) pre-coated with mAbAN 18 (Mabtech) and stimulated for 18 to 20 h with ⁇ g/ml Pb9 peptide (Kd-restricted CD8+ peptide SYIPSAEKI, a components of ME- TRAP) (Invitrogen, United Kingdom) or ⁇ g/ml EGFP peptide.
  • Cell concentrations were 2.5x10 6 /ml (for Pb9) or 5x10 6 /ml (for EGFP peptide).
  • a series of vectors were constructed expressing Photinus luciferase under the control of a series of promoters.
  • Some promoters contained natural or synthetic splice signals (Table 1 ). These included the human CMV immediate early cassette 94 promoter (hCMV), the human EF1 a promoter (hEF1 ), the simian CMV immediate early 94 promoter (sCMV), and the Xenopus EF1 a promoter.
  • Some constructs also contained the SV40 enhancer. A schematic organisation of the various promoters tested is shown in Table 1. Numbers before each construct are plasmid identifiers.
  • the activities shown are mean and standard deviation of three replicate transfections, in which 200ng of plasmid was co-transfected with 20ng of pGL4.74 (Promega, which expresses Renilla luciferase under the control of the TK promoter), and luciferase measurement were made with a Dual Luciferase assay (Promega).
  • Transient transfection of reporter constructs shows the Xenopus EF1 a promoter (xEF1 ) to be active in HeLa and 3T3 cells ( Figure 1 ).
  • Activity in vitro is significantly enhanced by the presence of the SV40 enhancer, and is comparable to that of the simian CMV promoter ( Figure 1 ).
  • adenoviruses were constructed using two of these promoter arrangements ( Figure 2).
  • Viruses containing the xEF1 -alpha cassette are stable, but those containing sCMV are not stable
  • AdC63 adenoviruses expressing the METRAP antigen, and derivatives containing a second expression cassette (described above) were used to infect were recovered following growth in 293 cells.
  • 293 cells were infected with AdC63 expressing METRAP, or METRAP together with EGFP under the control of the sCMV promoter, xEF1 promoter, or xEF1 promoter-SV40 enhancer at an MOI of 0.1. Under these conditions, virus is limiting, and infection of a single cell is likely to originate from one virion.
  • Immunofluorescence microscopy was performed 24 hours after infection.
  • Figure 2 shows representative cells from a large number examined; both METRAP and EGFP expression was demonstrable (Figure 4) in all cells, demonstrating function of the promoter, and viral stability.
  • the product of the xEF1 alpha cassette is immunogenic

Landscapes

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

Abstract

There is provided a viral vector comprising a first promoter sequence, wherein the first promoter sequence comprises: (a) a nucleic acid sequence having at least 70% sequence identity to the nucleic acid sequence of SEQ ID NO: 1;or (b) at least two fragments of (a), wherein each fragment comprises at least consecutive nucleotides of (a); or (c) a fragment of (a), wherein said fragment comprises at least 20 consecutive nucleotides of (a). Also provided are corresponding uses of the viral vector in medicine.

Description

Promoter sequence for DNA and viral vectors
This patent application claims priority to GB1004143.2 filed on 12 March 2010, which is hereby incorporated by reference in its entirety.
The present invention relates to viral vectors, to methods of making a viral vector, to host cells for use in making viral vectors, and to methods of expressing a gene or genes in a target cell using a single viral vector. Viral vectors are used in multiple applications in both basic biochemical research and medicine. Such vectors may be engineered using recombinant nucleic acid technology and are used to transfer a gene or genes of interest into a target cell, leading to the expression of the gene or genes and the production of the gene product or products encoded. Viral vectors have found application in vaccines against infectious diseases, in the treatment of cancer, and in gene therapy.
Expression of a gene from a viral vector requires that the gene has an associated promoter sequence. A promoter sequence is a length of DNA sequence to which RNA polymerase binds in order to begin the process of transcribing the coding sequence of the gene to RNA.
A number of promoters from different sources have been used in viral vectors, including the human cytomegalovirus (CMV) promoter, human telomerase promoters, a prostate specific promoter, the simian virus 40 (SV40) promoter, and the adenoviral major late and Iva2 promoters.
The need to insert the promoter into the genome of the viral vector creates an upper limit to the size of the promoter, particularly for smaller viruses, such as adenoviruses, retroviruses and lentiviruses. Smaller promoters are therefore preferable, as they leave more space in the viral genome for the gene or genes of interest. In addition, a promoter should be highly active in order to achieve high levels of transcription in the target cell.
In certain circumstances, it may be desirable to express multiple genes from a single viral vector. Examples of applications where such expression of multiple genes is desirable or essential include the expression of an adjuvant together with an antigen; the expression of multiple antigens, with one example being the need to express antigens from different pathogen serotypes to develop vaccines with broad coverage; the generation of immune responses against multiple targets; and the expression of a marker gene.
However, there are inherent difficulties in achieving the expression of multiple genes from a single viral vector, and while a number of methods are known in the art, none is satisfactory. One method has been to use a single cistron which expresses two proteins as a fusion protein. However, the use of a self-protein as part of such a fusion has been shown to lead to the development of an autoimmune response, which could potentially prove dangerous [Tenbusch, M., et al., BMC Immunol, 2008. 9: p. 13.] Expression of genes using internal ribosome entry sites has been attempted, but expression downstream of such sites may be weak [Gonzalez-Nicolini, V. and M. Fussenegger, Methods Mol Biol, 2008. 434: p. 221 -37. Cayer, MP., et al., J Immunol Methods, 2007. 322(1 -2): p. 1 18-27.]. The use of homologous promoters can be compromised by recombination within the viral genome in some configurations [Vogels, R., et al., J Gen Virol, 2007. 88(Pt 1 1 ): p. 2915-24. Belousova, N., et al., Mol Pharmacol, 2006. 70(5): p. 1488-93.].
There is therefore a need for a small and strong promoter, suitable for the expression of proteins in both single- and multi-cistronic viral vectors. The present invention solves one or more of the above technical problems by providing a viral vector comprising a first promoter sequence, wherein the first promoter sequence comprises:
(a) a nucleic acid sequence having at least 70% sequence identity to the nucleic acid sequence of SEQ ID NO: 1 ; or
(b) at least two fragments of (a), wherein each fragment comprises at least 10 consecutive nucleotides of (a); or
(c) a fragment of (a), wherein said fragment comprises at least 20 consecutive nucleotides of (a).
The present inventors have shown that a promoter based on the EF1 -alpha gene of Xenopus laevis (henceforth referred to as xEF1 -alpha = SEQ ID NO: 1 ) is unexpectedly suitable for use in viral vectors. SEQ ID NO: 1 excludes the presence of any known enhancer elements.
In one embodiment, the first promoter sequence consists of or comprises a nucleic acid sequence having at least 70% (such as at least 75, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or 100%) nucleic acid sequence identity to the nucleic acid sequence of SEQ ID NO: 1 .
In one embodiment, in the context of the first promoter sequence, the nucleic acid sequence identity with SEQ ID NO: 1 exists over a region of the nucleic acid sequence of SEQ ID NO: 1 that is at least 100 consecutive nucleotides in length (e.g. at least 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, consecutive nucleotides or the entire nucleotide in length).
In one embodiment, in the context of the 'at least two fragment' aspect of the first promoter sequence, each of said fragments comprises (or consists of) at least 10 consecutive nucleotides (e.g. at least 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 1 10, 1 15, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180 or 185 nucleotides). In one embodiment, the at least two fragments of (a) may number at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9 or at least 10 fragments. In one embodiment, the overall nucleotide length of the 'at least two fragment aspect' is at least 100 nucleotides, or at least 150 nucleotides, or at least 200 nucleotides, or at least 250 nucleotides, or at least 300 nucleotides, or at least 350 nucleotides in length.
In one embodiment, in the context of the 'at least two fragment' aspect (b) of the first promoter sequence, a fragment may be selected from a sequence having least 70% (such as at least 75, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or 100%) nucleic acid sequence identity to any one or more of the following SEQ ID NO: 1 nucleic acid fragments:
18-27; 34-47; 41 -50; 83-92; 84-93; 93-103; 124-133; 158-167; 159-169; 165-174; 176-185; 177-186; 178-187; 176-185; 177-186; 178-187; 195-204; 198-207; 200- 209; 202-21 1 ; 210-221 ; 221 -230; 226-235; 232-241 ; 237-246; 232-241 ; 237-246; 250-259; 261 -270; 265-274; 269-278; 273-282; 327-336; 337-346; 342-351 ; 344- 353; and/ or 345-354.
In one embodiment, in the context of the fragment aspect (c) of the first promoter sequence, said fragment comprises (or consists of) at least 21 consecutive nucleotides (e.g. at least 24, 30, 42, 51 , 60, 72, 81 , 90, 102, 1 1 1 , 120, 132, 141 , 150, 162, 171 , 180, 192, 201 , 210, 222, 231 , 240, 252, 261 , 270, 282, 291 , 300, 312, 321 , 330, 342, 351 , 360 or 372 nucleotides) of nucleic acid sequence according to aspect (a). In one embodiment, the fragment aspect (c) may include any one or more of the 'at least two' fragments described immediately above for aspect (b).
In one embodiment, in the context of the first promoter sequence, the fragment (either aspect (b) or aspect (c)) has a sequence length that is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of that of the sequence of the full-length nucleic acid sequence SEQ ID NO: 1 . In one embodiment, in the context of the fragment aspect of first promoter sequence (either aspect (b) or aspect (c)), the nucleic acid sequence identity with SEQ ID NO: 1 exists over a region of the nucleic acid sequence of SEQ ID NO: 1 that is at least 20 consecutive nucleotides in length. In one embodiment, the region of identity is over a length of SEQ ID NO: 1 that is the same length as the length of the fragment of the invention. In another embodiment, the region of identity is over a length of SEQ ID NO: 1 that is at least 125, 150, 175, 200, 225, 250, 275, 300, 325, 350 or 375 consecutive nucleotides of SEQ ID NO: 1. As used herein, the term "viral vector" means any recombinant virus suitable for introducing genetic material into a cell.
In one embodiment, the first promoter sequence provides a strong promoter, capable of facilitating a high level of transcription. In one embodiment, the first promoter sequence is of a size advantageous for use in a viral vector such as an adenoviral vector. In one embodiment, the first promoter sequence will not undergo homologous recombination with the human genome. In this regard, a regulatory consideration, when viruses are used in humans, is the potential risk of homologous integration of expression cassettes containing human promoters into the human genome. For this reason, promoters lacking primary sequence homology with human sequences may be preferable for regulatory reasons.
In one embodiment, the viral vector further comprises an enhancer sequence; wherein the enhancer sequence increases transcription from the first promoter sequence.
As used herein, the term "enhancer sequence" means any nucleic acid sequence, the presence of which increases transcription from the first promoter sequence (for example, by leading to an increase in the number of transcripts produced over a given period of time, in comparison to the number of transcripts produced in the same period of time in the absence of the enhancer). The enhancer sequence may be located anywhere in the viral vector, and may be present in either orientation.
In one embodiment, the enhancer sequence comprises a nucleic acid sequence having at least 70% identity to the nucleic acid sequence of SEQ ID NO: 2, or a fragment thereof comprising at least 10 consecutive nucleotides thereof. The nucleic acid sequence represented by SEQ ID NO: 2 is based on the enhancer sequence of SV40 virus (Simian Virus 40). In one embodiment, the enhancer sequence consists of or comprises a nucleic acid sequence having at least 70% (such as at least 75, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or 100%) nucleic acid sequence identity to the nucleic acid sequence of SEQ ID NO: 2, or a fragment thereof comprising at least 10 consecutive nucleotides thereof.
In one embodiment, in the context of the enhancer sequence, the nucleic acid sequence identity exists over a region of the nucleic acid sequence that is at least 10 consecutive nucleotides in length (e.g. at least 15, 25, 30, 35, 40, 45, 50, or 55 consecutive nucleotides in length).
In the context of the enhancer sequence, a fragment comprises (or consists of) at least 10 consecutive nucleotides of said nucleic acid sequence (e.g. at least 15, 20, 25, 30, 35, 40, 42, 44, 46, 48, 50, 52 or 54 consecutive nucleotides thereof). In one embodiment, in the context of the enhancer sequence, a fragment of a nucleic acid sequence has a sequence length that is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of that of the sequence of the full-length nucleic acid sequence. In one embodiment, the enhancer sequence comprises or consists of a nucleic acid sequence having at least 70% (such as at least 75, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or 100%) sequence identity to the nucleic acid sequence of SEQ ID NO: 3, or a fragment thereof comprising at least 10 consecutive nucleotides thereof. The nucleic acid sequence represented by SEQ ID NO: 3 is based on the Xenopus laevis EF1 -alpha gene enhancer sequence.
In one embodiment, in the context of the enhancer sequence, the nucleic acid sequence identity exists over a region of the nucleic acid sequence that is at least 10 consecutive nucleotides in length (e.g. at least 15, 25, 30, 35, 40, 45, 50, or 55 consecutive nucleotides in length).
In the context of the enhancer sequence, a fragment comprises (or consists of) at least 10 consecutive nucleotides of said nucleic acid sequence (e.g. at least 15, 20, 25, 30, 35, 40, 42, 44, 46, 48, 50, 52 or 54 consecutive nucleotides thereof). In one embodiment, in the context of the enhancer sequence, a fragment of a nucleic acid sequence has a sequence length that is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of that of the sequence of the full-length nucleic acid sequence. In one embodiment, the viral vector further comprises at least one gene; wherein the first promoter sequence is operably-linked to the at least one gene, and facilitates the expression thereof.
As used herein, the term "gene" encompasses both protein-coding and non protein-coding genes. Thus, in one embodiment, the viral vector comprises at least one protein-coding gene. In one embodiment, the viral vector comprises at least one non protein-coding gene. The non protein-coding gene may encode an RNA. Thus, in one embodiment, the non protein-coding gene encodes a small interfering RNA (siRNA). Genes suitable for use in the present invention include, but are not limited to, those coding for the following: antigens, adjuvants (including viral, bacterial and eukaryotic gene products), gene products expressed by tumours or cancers, anti- tumour / anti-cancer gene products, enzymes, reporters, and receptors.
An example of an antigen suitable for use in the present invention is the METRAP antigen. Thus, in one embodiment, the gene encodes the METRAP antigen. An example of an adjuvant suitable for use in the present invention is granulocyte-macrophage colony-stimulating factor (GM-CSF). Thus, in one embodiment, the gene encodes GM-CSF.
An example of a gene product expressed by a tumour or cancer suitable for use in the present invention is a tumour-specific antigen. In this regard, a tumour- specific antigen is a protein or molecule which is either unique to a tumour cell or is present in the tumour cell in much greater abundance compared to a non- tumour cell. It is evident from the above that any gene can be expressed using the viral vector of the present invention, and that there is no intention to limit the present invention to viral vectors expressing any one particular gene. A skilled person will be familiar with other enzyme, reporter and receptor genes which would be suitable for use in the present invention.
As used herein, "operably-linked" means that the nucleic acid sequences being linked are arranged so that they function in concert for their intended purposes - for example, transcription initiates in the promoter and proceeds through the coding polynucleotide segment to the terminator. Thus, in one embodiment, transcription may be initiated in the first promoter sequence and proceed through the coding sequence of the at least one gene, leading to the expression of said gene. In one embodiment, the viral vector further comprises a second promoter sequence; wherein the first promoter sequence and the second promoter sequence are different.
In the context of the second promoter sequence, "different" means that the second promoter sequence is any promoter sequence that is not the above- defined first promoter sequence. Thus, in one embodiment, the second promoter sequence is a promoter sequence lacking sequence homology with the first promoter sequence, to the extent that homologous recombination between the first and second promoter sequences cannot take place.
In one embodiment, the first promoter sequence is operably-linked to a first gene, and facilitates the expression thereof; and the second promoter sequence is operably-linked to a second gene, and facilitates the expression thereof. Thus, in one embodiment, the viral vector is a bicistronic viral vector.
In one embodiment, the viral vector comprises at least one additional promoter sequence operably-linked to an additional gene, wherein the additional promoter sequence facilitates the expression of the additional gene. Thus, the viral vector may comprise a third promoter sequence, a fourth promoter sequence, or a fifth promoter sequence, or additional promoter sequences beyond a fifth promoter sequence, each additional promoter sequence operably-linked to a gene and facilitating the expression thereof. Thus, in one embodiment, the viral vector is a multicistronic vector. In one embodiment, the second promoter sequence is selected from the group consisting of: human CMV promoter, simian CMV promoter, murine or other mammalian CMV promoters, promoters derived from other herpes viruses, promoters from other types of virus, and mammalian promoters.
In one embodiment, the viral vector is any virus which can serve as a viral vector. Suitable viruses are those which infect the species of interest, can be propagated in vitro, and can be modified by recombinant nucleotide technology known in the art.
In one embodiment, the viral vector is selected from the group consisting of: an adenovirus vector; an adeno-associated virus vector; a pox virus vector, such as a fowlpox virus vector; an alpha virus vector; a bacloviral vector; a herpes virus vector; a retrovirus vector, such as a lentivirus vector; a Modified Vaccinia virus Ankara vector; a Ross River virus vector; a Sindbis virus vector; a Semliki Forest virus vector; and a Venezuelan Equine Encephalitis virus vector.
In one embodiment, the viral vector is an adenovirus vector. Thus, in one embodiment, the viral vector is an adenovirus vector comprising a first promoter sequence, the first promoter sequence being as defined above. In one embodiment, the viral vector further comprises at least one repressor binding site sequence. The repressor binding site sequence may be introduced into the viral vector by recombinant DNA technology. The repressor binding site sequence may be associated with the first promoter sequence such that it is able to control the expression of a gene operably-linked to the first promoter sequence. In this regard, binding of a repressor protein to the repressor binding site enables negative regulation of the first promoter sequence, decreasing or suppressing expression of a gene operably-linked to said promoter. In one embodiment, the repressor protein is produced by the host cell in which the viral vector of the present invention is grown. An example of a suitable repressor binding site sequence is the tetracycline operator site sequence. This sequence is able to bind tetracycline repressor proteins, so decreasing gene expression from an associated promoter sequence. Thus, in one embodiment, the viral vector includes an artificial repressible promoter sequence. Other, non-limiting, examples of suitable repressor binding site sequences are: a Lac operator, a Mutant Lac operator, and a LexA operator.
In one embodiment, the viral vector further comprises an additional repressor binding site sequence for each additional promoter sequence beyond the first promoter sequence.
In one embodiment, the at least one repressor binding site sequence comprises a nucleic acid sequence having at least 70% sequence identity to the nucleic acid sequence of SEQ ID NO: 4, or a fragment thereof comprising at least 10 consecutive nucleotides thereof. The nucleic acid sequence represented by SEQ ID NO: 4 is based on the tetracycline operator site. In one embodiment, the viral vector further comprises two tetracycline operator sites.
In one embodiment, the at least one repressor binding site sequence comprises or consists of a nucleic acid sequence having at least 70% (such as at least 75, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or 100%) nucleic acid sequence identity to the nucleic acid sequence of SEQ ID NO: 4, or a fragment thereof comprising at least 10 consecutive nucleotides thereof. In one embodiment, the nucleic acid sequence identity exists over a region of the nucleic acid sequence that is at least 10 consecutive nucleotides in length (e.g. at least 15, 25, 30, 35, 40, 45, 50, or 55 consecutive nucleotides in length).
In the context of the at least one repressor binding site sequence, a fragment comprises (or consists of) at least 10 consecutive nucleotides of said nucleic acid sequence (e.g. at least 15, 20, 25, 30, 35, 40, 42, 44, 46, 48, 50, 52 or 54 consecutive nucleotides thereof).
In one embodiment, in the context of the at least one repressor binding site sequence, a fragment of a nucleic acid sequence has a sequence length that is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of that of the sequence of the full-length nucleic acid sequence.
In one embodiment, the viral vector comprises at least one repressor binding site sequence (for example a tetracycline operator site) inserted downstream of the TATA box of the first promoter sequence. The number of repressor binding sites (e.g. the tetracycline operator) may be two, or a number greater than two. Thus, in one embodiment, the viral vector comprises two repressor binding sites (e.g. the tetracycline operator) inserted downstream of the TATA box of the first promoter sequence.
In the context of a tetracycline operator site sequence, a tetracycline operator site is able to bind the tetracycline repressor protein; the binding of this protein to the operator site acts to inhibit transcription from the proximal promoter. In the presence of tetracycline, the tetracycline repressor protein is no longer bound to the operator sequence and transcription from the proximal promoter sequence is no longer repressed.
In one embodiment, the viral vector comprises at least one repressor binding site sequence (for example a tetracycline operator site) inserted downstream of the TATA box of the second promoter sequence. The number of repressor binding sites (e.g. the tetracycline operator) may be two, or a number greater than two. Thus, in one embodiment, the viral vector comprises two repressor binding sites (e.g. the tetracycline operator) inserted downstream of the TATA box of the second promoter sequence. In one aspect, the invention provides a method of making a viral vector, comprising providing a nucleic acid, wherein the nucleic acid comprises a nucleic acid sequence encoding a viral vector (as described above); transfecting a host cell with the nucleic acid; culturing the host cell under conditions suitable for the expression of the nucleic acid; and obtaining the viral vector from the host cell.
The nucleic acid comprising a sequence encoding a viral vector (as described above) may be generated by the use of any technique for manipulating and generating recombinant nucleic acid known in the art. As used herein, "transfecting" may mean any non-viral method of introducing nucleic acid into a cell. The nucleic acid may be any nucleic acid suitable for transfecting a host cell. Thus, in one embodiment, the nucleic acid is a plasmid. The host cell may be any cell in which a viral vector (as described above) may be grown. As used herein, "culturing the host cell under conditions suitable for the expression of the nucleic acid" means using any cell culture conditions and techniques known in the art which are suitable for the chosen host cell, and which enable the viral vector to be produced in the host cell. As used herein, "obtaining the viral vector", means using any technique known in the art that is suitable for separating the viral vector from the host cell. Thus, in one embodiment, the host cells are lysed to release the viral vector. The viral vector may subsequently be isolated and purified using any suitable method or methods known in the art.
In one aspect, the invention provides a host cell, comprising a viral vector (as described above). The host cell may be any cell in which a viral vector (as described above) may be grown. Thus, in one embodiment, the host cell is selected from the group consisting of: a 293 cell, a CHO cell, a CCL81.1 cell, a Vera cell, a HELA cell, a Per.C6 cell, and a BHK cell.
In one aspect, the invention provides a method of expressing in a target cell at least one protein, comprising providing a viral vector (as described above); and introducing the viral vector into a target cell. The method of expressing in a target cell at least one protein may be carried out on a target cell in vitro, in vivo or ex vivo. Thus, the target cell may be part of an in vitro cell culture, or part of a subject in vivo, or part of an ex vivo organ or tissue. In one embodiment, the invention provides an in vitro method of expressing in a target cell at least one protein, comprising providing a viral vector (as described above); and introducing the viral vector into a target cell. In one embodiment, the viral vector (as described above) is provided according to the method of making a viral vector described above. The viral vector may be introduced into the target cell according to any suitable method known in the art.
The viral vector of the present invention has multiple utilities, which are described below. In one aspect, the invention provides a viral vector (as described above) for use in medicine.
In one aspect, the invention provides a viral vector (as described above), for use in gene therapy. Methods and applications of gene therapy are known in the art. In one embodiment, the viral vector is for use in any method which aims to prevent, treat or cure a disease or diseases through the introduction of novel genetic material into the cells of a subject.
In one aspect, the invention provides a viral vector for use in the treatment of cancer. In one embodiment the viral vector (as described above), for use in the treatment of cancer may comprise at least one gene encoding a tumour-specific antigen (such tumour-specific antigen being as described above). The tumour- specific antigen may be displayed on the external surface of the tumour cell, or may be present internally. In one embodiment, the viral vector (as described above), for use in the treatment of cancer, is used wherein the treatment comprises administering the viral vector to a subject and stimulating in the subject an immunogenic response against a tumour cell or cells present in the subject. Thus, the subject's immune system may be stimulated to attack a tumour present in the subject. In one embodiment, the viral vector (as described above), for use in the treatment of cancer, may comprise at least one gene encoding a gene product which has anti-tumour or anti-cancer properties. In this regard, the gene product may be a nucleic acid (for example, a small interfering RNA) or a protein. The gene product may inhibit the proliferation and/or division of tumour/cancer cells. Alternatively or additionally, the gene product may cause the death of tumour/cancer cells (for example, via necrosis or apoptosis).
In one aspect, the invention provides a viral vector (as described above), for use in the treatment of allergy. In one embodiment, the viral vector (as described above), for use in the treatment of allergy, may further comprise a gene encoding an allergen. In this regard, the viral vector (as described above), for use in the treatment of allergy, may be used wherein the treatment of allergy comprises a prophylactic treatment to prevent the development of an allergy in a subject, or a therapeutic treatment to reduce or abolish the symptoms of an allergy in a subject.
In one aspect, the invention provides a viral vector (as described above), for use in stimulating or inducing an immune response in a subject. In one embodiment, stimulating or inducing an immune response in a subject comprises administering to the subject a viral vector (as described above).
In one embodiment, the viral vector (as described above), for use in stimulating or inducing an immune response in a subject, comprises at least one gene encoding an antigen. In one embodiment, the antigen is the METRAP antigen. In one embodiment, the viral vector (as described above), for use in stimulating or inducing an immune response in a subject, comprises at least one gene encoding an adjuvant. In one embodiment, the viral vector (as described above), for use in stimulating or inducing an immune response in a subject, comprises a first gene encoding an antigen, and a second gene encoding an adjuvant. In one embodiment, stimulating or inducing an immune response in a subject comprises administering a viral vector (as described above) to a subject, wherein said viral vector is administered substantially prior to, simultaneously with or subsequent to another immunogenic composition. In one aspect, the invention provides a viral vector (as described above), for use in the treatment or prevention of at least one infectious disease. In one embodiment, the at least one infectious disease is selected from the group consisting of diseases caused by: Plasmodia, influenza viruses, Mycobacterium tuberculosis, Mycobacterium bovis, other Mycobacteria, hepatitis C virus, other flaviviruses, hepatitis B virus, human immunodeficiency virus, other retroviruses, Staphylococcus aureus, other Staphylococci, Streptococcus pneumoniae, Streptococcus pyogenes, other Streptococci, Haemophilus influenzae, Neisseria meningitides. In one embodiment, the treatment or prevention of at least one infectious disease comprises administering to a subject a viral vector (as described above) wherein said viral vector is administered substantially prior to, simultaneously with or subsequent to another immunogenic composition. Prior, simultaneous and sequential administration regimes are discussed in more detail below.
The viral vector of the present invention may be useful for inducing a range of immune responses and may therefore be useful in methods for treating a range of diseases. As used herein, the term "treatment" or "treating" embraces therapeutic or preventative/prophylactic measures, and includes post-infection therapy and amelioration of an infectious disease. As used herein, the term "preventing" includes preventing the initiation of an infectious disease and/or reducing the severity or intensity of an infectious disease.
A viral vector of the invention (as described above) may be administered to a subject (typically a mammalian subject such as a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) already having an infectious disease, to treat or prevent said infectious disease. In one embodiment, the subject is suspected of having come into contact with an infectious disease (or the disease-causing agent), or has had known contact with an infectious disease (or the disease-causing agent), but is not yet showing symptoms of exposure to said infectious disease (or said disease-causing agent).
When administered to a subject (e.g. a mammal such as a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) that already has an infectious disease, or is showing symptoms associated with an infectious disease, a viral vector of the invention (as described above) can cure, delay, reduce the severity of, or ameliorate one or more symptoms of, the infectious disease; and/or prolong the survival of a subject beyond that expected in the absence of such treatment.
Alternatively, a viral vector of the invention (as described above) may be administered to a subject (e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) who may ultimately contract an infectious disease, in order to prevent, cure, delay, reduce the severity of, or ameliorate one or more symptoms of, said infectious disease; or in order to prolong the survival of a subject beyond that expected in the absence of such treatment. In one embodiment, the subject has previously been exposed to an infectious disease. For example, the subject may have had an infectious disease in the past (but is optionally not currently infected with the disease-causing agent of the infectious disease). The subject may be latently infected with an infectious disease. Alternatively, or in addition, the subject may have been vaccinated against said infectious disease in the past.
The treatments and preventative therapies in which viral vectors of the present invention may be used are applicable to a variety of different subjects of different ages. In the context of humans, the therapies are applicable to children (e.g. infants, children under 5 years old, older children or teenagers) and adults. In the context of other animal subjects (e.g. mammals such as bovine, porcine or equine subjects), the therapies are applicable to immature subjects (e.g. calves, piglets, foals) and mature/adult subjects. The treatments and preventative therapies of the present invention are applicable to subjects who are immunocompromised or immunosuppressed (e.g. human patients who have HIV or AIDS, or other animal patients with comparable immunodeficiency diseases), subjects who have undergone an organ transplant, bone marrow transplant, or who have genetic immunodeficiencies.
The viral vectors of the invention (as described above) can be employed as vaccines. As used, herein, a "vaccine" is a formulation that, when administered to an animal subject such as a mammal (e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) stimulates a protective immune response against an infectious disease. The immune response may be a humoral and/or a cell-mediated immune response. Thus, the vaccine may stimulate B- cells and/or T-cells. A vaccine of the invention can be used, for example, to protect an animal from the effects of an infectious disease (for example, malaria, influenza or tuberculosis).
The term "vaccine" is herein used interchangeably with the terms "therapeutic/prophylactic composition", "formulation", "antigenic composition", or "medicament".
In one aspect, the invention provides a vaccine composition, comprising a viral vector (as described above); and a pharmaceutically acceptable carrier.
The vaccine of the invention (as defined above) in addition to a pharmaceutically acceptable carrier can further be combined with one or more of a salt, excipient, diluent, adjuvant, immunoregulatory agent and/or antimicrobial compound. In one aspect, the invention provides an immunological composition, comprising a viral vector (as described above); and a pharmaceutically acceptable carrier.
The immunological composition in addition to a pharmaceutically acceptable carrier can further be combined with one or more of a salt, excipient, diluent, adjuvant, immunoregulatory agent and/or antimicrobial compound.
In one aspect, the invention provides a pharmaceutical composition, comprising a viral vector (as described above); and a pharmaceutically acceptable carrier. The pharmaceutical composition in addition to a pharmaceutically acceptable carrier can further be combined with one or more of a salt, excipient, diluent, adjuvant, immunoregulatory agent and/or antimicrobial compound.
The viral vector may be formulated into a vaccine, immunogenic composition or pharmaceutical composition as neutral or salt forms. Pharmaceutically acceptable salts include acid addition salts formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or with organic acids such as acetic, oxalic, tartaric, maleic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
Administration of immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) is generally by conventional routes e.g. intravenous, subcutaneous, intraperitoneal, or mucosal routes. The administration may be by parenteral administration; for example, a subcutaneous or intramuscular injection.
Accordingly, immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention may be prepared as injectables, either as liquid solutions or suspensions. Solid forms suitable for solution in, or suspension in, liquid prior to injection may alternatively be prepared. The preparation may also be emulsified, or the peptide encapsulated in liposomes or microcapsules.
The active ingredients are often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like and combinations thereof. In addition, if desired, the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, and/or pH buffering agents. Vaccine and immunological compositions of the present invention may comprise adjuvants which enhance the effectiveness of the vaccine or immunological composition. Generally, the carrier is a pharmaceutically-acceptable carrier. Non-limiting examples of pharmaceutically acceptable carriers include water, saline, and phosphate-buffered saline. In some embodiments, however, the composition is in lyophilized form, in which case it may include a stabilizer, such as bovine serum albumin (BSA). In some embodiments, it may be desirable to formulate the composition with a preservative, such as thiomersal or sodium azide, to facilitate long term storage.
Examples of additional adjuvants which may be effective include but are not limited to: complete Freunds adjuvant (CFA), Incomplete Freunds Adjuvant (IVA), Saponin, a purified extract fraction of Saponin such as Quil A, a derivative of Saponin such as QS-21 , lipid particles based on Saponin such as ISCOM/ISCOMATIX, E. coli heat labile toxin (LT) mutants such as LTK63 and/or LTK72, aluminium hydroxide, N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr- MDP), N-acetyl-nor-muramyl-L-alanyl-D-isoglutamine (CGP 1 1637, referred to as nor-MDP), N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1 '-2'- dipalmitoyl-sn-glycero-3-hydroxyphosphoryl oxy)-ethylamine (CGP 19835A, referred to as MTP-PE), and RIBI, which contains three components extracted from bacteria, monophosphoryl lipid A, trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2 % squalene/Tween 80 emulsion.
Examples of buffering agents include, but are not limited to, sodium succinate (pH 6.5), and phosphate buffered saline (PBS; pH 6.5 and 7.5). Additional formulations which are suitable for other modes of administration include suppositories and, in some cases, oral formulations or formulations suitable for distribution as aerosols. For suppositories, traditional binders and carriers may include, for example, polyalkylene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in the range of 0.5% to 10%, preferably 1 %-2%. Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders.
It may be desired to direct the viral vectors of the present invention (as described above) to the respiratory system of a subject; for example, for use in the treatment or prevention of a respiratory disease, or for the targeting of a gene therapy to the respiratory system (such as to the lungs). Efficient transmission of a therapeutic/prophylactic composition or medicament to the site of infection in the lungs may be achieved by oral or intra-nasal administration.
Formulations for intranasal administration may be in the form of nasal droplets or a nasal spray. An intranasal formulation may comprise droplets having approximate diameters in the range of 100-5000 μηη, such as 500-4000 μηη, 1000-3000 μηη or 100-1000 μηη. Alternatively, in terms of volume, the droplets may be in the range of about 0.001 -100 μΙ, such as 0.1 -50 μΙ or 1 .0-25 μΙ, or such as 0.001 -1 μΙ.
Alternatively, the therapeutic/prophylactic formulation or medicament may be an aerosol formulation. The aerosol formulation may take the form of a powder, suspension or solution. The size of aerosol particles is relevant to the delivery capability of an aerosol. Smaller particles may travel further down the respiratory airway towards the alveoli than would larger particles. In one embodiment, the aerosol particles have a diameter distribution to facilitate delivery along the entire length of the bronchi, bronchioles, and alveoli. Alternatively, the particle size distribution may be selected to target a particular section of the respiratory airway, for example the alveoli. In the case of aerosol delivery of the medicament, the particles may have diameters in the approximate range of 0.1 - 50 μηη, preferably 1 -25 μηη, more preferably 1 -5 μηη. Aerosol particles may be for delivery using a nebulizer (e.g. via the mouth) or nasal spray. An aerosol formulation may optionally contain a propellant and/or surfactant.
By controlling the size of the droplets/particles to within the defined range of the present invention, it is possible to avoid (or minimize) inadvertent medicament delivery to the alveoli and thus avoid alveoli-associated pathological problems such as inflammation and fibrotic scarring of the lungs.
Intra-nasal vaccination engages both T- and B-cell mediated effector mechanisms in nasal and bronchus associated mucosal tissues, which differ from other mucosa-associated lymphoid tissues. The protective mechanisms invoked by the intranasal route of administration may include: the activation of T- lymphocytes with preferential lung homing; up-regulation of co-stimulatory molecules (e.g. B7.2); and/or activation of macrophages or secretory IgA antibodies.
Intranasal delivery of viral vectors of the invention (as described above) may facilitate the invoking of a mucosal antibody response, which is favoured by a shift in the T-cell response toward the Th2 phenotype which helps antibody production. A mucosal response is characterised by enhanced IgA production, and a Th2 response is characterised by enhanced IL-4 production. In one embodiment, the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention comprise a pharmaceutically acceptable carrier, and optionally one or more of a salt, excipient, diluent and/ or adjuvant. In one embodiment, the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention may comprise one or more immunoregulatory agents selected from, for example, immunoglobulins, antibiotics, interleukins (e.g. IL-2, IL-12), and/or cytokines (e.g. IFNy). In one embodiment, the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention may comprise one or more antimicrobial compounds, (for example, conventional anti-tuberculosis drugs such as rifampicin, isoniazid, ethambutol or pyrizinamide).
The immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention may be given in a single dose schedule (i.e. the full dose is given at substantially one time). Alternatively, the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention may be given in a multiple dose schedule.
A multiple dose schedule is one in which a primary course of treatment (e.g. vaccination) may be with 1 -6 separate doses, followed by other doses given at subsequent time intervals required to maintain and or reinforce the immune response, for example (for human subjects), at 1 -4 months for a second dose, and if needed, a subsequent dose(s) after a further 1 -4 months. The dosage regimen will be determined, at least in part, by the need of the individual and be dependent upon the judgment of the practitioner (e.g. doctor or veterinarian).
Simultaneous administration means administration at (substantially) the same time. Sequential administration of two or more compositions/therapeutic agents/vaccines means that the compositions/therapeutic agents/vaccines are administered at (substantially) different times, one after the other. For example, in one embodiment, the vaccine of the present invention may be administered as part of a 'prime-boost' vaccination regime.
In one embodiment, the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention can be administered to a subject such as a mammal (e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) in conjunction with (simultaneously or sequentially) one or more immunoregulatory agents selected from, for example, immunoglobulins, antibiotics, interleukins (e.g. IL-2, IL-12), and/or cytokines (e.g. IFNy).
In one embodiment, the immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) of the invention can be administered to a subject such as a mammal (e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) in conjunction with (simultaneously or sequentially) one or more antimicrobial compounds, such as conventional anti-tuberculosis drugs (e.g. rifampicin, isoniazid, ethambutol or pyrizinamide).
The immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) may contain 5% to 95% of active ingredient, such as at least 10% or 25% of active ingredient, or at least 40% of active ingredient or at least 50, 55, 60, 70 or 75% active ingredient. The immunogenic compositions, therapeutic formulations, medicaments, pharmaceutical compositions, and prophylactic formulations (e.g. vaccines) are administered in a manner compatible with the dosage formulation, and in such amount as will be prophylactically and/or therapeutically effective.
In this regard, as used herein, an "effective amount" is a dosage or amount that is sufficient to achieve a desired biological outcome. As used herein, a "therapeutically effective amount" is an amount which is effective, upon single or multiple dose administration to a subject (such as a mammal - e.g. a human, bovine, porcine, ovine, caprine, equine, cervine, canine or feline subject) for treating, preventing, curing, delaying, reducing the severity of, ameliorating at least one symptom of a disorder or recurring disorder, or prolonging the survival of the subject beyond that expected in the absence of such treatment.
Accordingly, the quantity of active ingredient to be administered depends on the subject to be treated, capacity of the subject's immune system to generate a protective immune response, and the degree of protection desired. Precise amounts of active ingredient required to be administered may depend on the judgment of the practitioner and may be particular to each subject.
As used herein, the terms "nucleic acid sequence" and "polynucleotide" are used interchangeably and do not imply any length restriction. As used herein, the terms "nucleic acid" and "nucleotide" are used interchangeably. The terms "nucleic acid sequence" and "polynucleotide" embrace DNA (including cDNA) and RNA sequences. The polynucleotide sequences of the present invention include nucleic acid sequences that have been removed from their naturally occurring environment, recombinant or cloned DNA isolates, and chemically synthesized analogues or analogues biologically synthesized by heterologous systems. The polynucleotides of the present invention may be prepared by any means known in the art. For example, large amounts of the polynucleotides may be produced by replication in a suitable host cell. The natural or synthetic DNA fragments coding for a desired fragment will be incorporated into recombinant nucleic acid constructs, typically DNA constructs, capable of introduction into and replication in a prokaryotic or eukaryotic cell. Usually the DNA constructs will be suitable for autonomous replication in a unicellular host, such as yeast or bacteria, but may also be intended for introduction to and integration within the genome of a cultured insect, mammalian, plant or other eukaryotic cell lines.
The polynucleotides of the present invention may also be produced by chemical synthesis, e.g. by the phosphoramidite method or the triester method, and may be performed on commercial automated oligonucleotide synthesizers. A double- stranded fragment may be obtained from the single stranded product of chemical synthesis either by synthesizing the complementary strand and annealing the strand together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence.
When applied to a nucleic acid sequence, the term "isolated" in the context of the present invention denotes that the polynucleotide sequence has been removed from its natural genetic milieu and is thus free of other extraneous or unwanted coding sequences (but may include naturally occurring 5' and 3' untranslated regions such as promoters and terminators), and is in a form suitable for use within genetically engineered protein production systems. Such isolated molecules are those that are separated from their natural environment. In view of the degeneracy of the genetic code, considerable sequence variation is possible among the polynucleotides of the present invention. Degenerate codons encompassing all possible codons for a given amino acid are set forth below: Amino Acid Codons Degenerate Codon
Cys TGC TGT TGY
Ser AGC AGT TCA TCC TCG TCT WSN
Thr ACA ACC ACG ACT ACN
Pro CCA CCC CCG CCT CCN
Ala GCA GCC GCG GCT GCN
Gly GGA GGC GGG GGT GGN
Asn AAC AAT AAY
Asp GAC GAT GAY
Glu GAA GAG GAR
Gin CAA CAG CAR
His CAC CAT CAY
Arg AGA AGG CGA CGC CGG CGT MGN
Lys AAA AAG AAR
Met ATG ATG
He ATA ATC ATT ATH
Leu CTA CTC CTG CTT TTA TTG YTN
Val GTA GTC GTG GTT GTN
Phe TTC 1 1 1 TTY
Tyr TAC TAT TAY
Trp TGG TGG
Ter TAA TAG TGA TRR
Asn/ Asp RAY
Glu/ Gin SAR
Any NNN
One of ordinary skill in the art will appreciate that flexibility exists when determining a degenerate codon, representative of all possible codons encoding each amino acid. For example, some polynucleotides encompassed by the degenerate sequence may encode variant amino acid sequences, but one of ordinary skill in the art can easily identify such variant sequences by reference to the amino acid sequences of the present invention.
A "variant" nucleic acid sequence has substantial homology or substantial similarity to a reference nucleic acid sequence (or a fragment thereof). A nucleic acid sequence or fragment thereof is "substantially homologous" (or "substantially identical") to a reference sequence if, when optimally aligned (with appropriate nucleotide insertions or deletions) with the other nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 70%, 75%, 80%, 82, 84, 86, 88, 90, 92, 94, 96, 98 or 99% of the nucleotide bases. Methods for homology determination of nucleic acid sequences are known in the art.
Alternatively, a "variant" nucleic acid sequence is substantially homologous with (or substantially identical to) a reference sequence (or a fragment thereof) if the "variant" and the reference sequence they are capable of hybridizing under stringent (e.g. highly stringent) hybridization conditions. Nucleic acid sequence hybridization will be affected by such conditions as salt concentration (e.g. NaCI), temperature, or organic solvents, in addition to the base composition, length of the complementary strands, and the number of nucleotide base mismatches between the hybridizing nucleic acids, as will be readily appreciated by those skilled in the art. Stringent temperature conditions are preferably employed, and generally include temperatures in excess of 30°C, typically in excess of 37°C and preferably in excess of 45°C. Stringent salt conditions will ordinarily be less than 1000 mM, typically less than 500 mM, and preferably less than 200 mM. The pH is typically between 7.0 and 8.3. The combination of parameters is much more important than any single parameter.
One of ordinary skill in the art appreciates that different species exhibit "preferential codon usage". As used herein, the term "preferential codon usage" refers to codons that are most frequently used in cells of a certain species, thus favouring one or a few representatives of the possible codons encoding each amino acid. For example, the amino acid threonine (Thr) may be encoded by ACA, ACC, ACG, or ACT, but in mammalian host cells ACC is the most commonly used codon; in other species, different Thr codons may be preferential. Preferential codons for a particular host cell species can be introduced into the polynucleotides of the present invention by a variety of methods known in the art. Introduction of preferential codon sequences into recombinant DNA can, for example, enhance production of the protein by making protein translation more efficient within a particular cell type or species.
Thus, in one embodiment of the invention, the nucleic acid sequence is codon optimized for expression in a host cell.
A "fragment" of a polynucleotide of interest comprises a series of consecutive nucleotides from the sequence of said full-length polynucleotide. By way of example, a "fragment" of a polynucleotide of interest may comprise (or consist of) at least 30 consecutive nucleotides from the sequence of said polynucleotide (e.g. at least 35, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800 850, 900, 950 or 1000 consecutive nucleic acid residues of said polynucleotide). A fragment may include at least one antigenic determinant and/or may encode at least one antigenic epitope of the corresponding polypeptide of interest.
Key to SEQ ID NOs.
SEQ ID NO: 1 Core EF1 -alpha promoter sequence from Xenopus laevis. SEQ ID NO: 2 Enhancer sequence from SV40.
SEQ ID NO: 3 Enhancer sequence from EF1 -alpha gene of Xenopus laevis. SEQ ID NO: 4 Tetracycline operator site sequence.
SEQ ID NO: 5 Full length sequence of Xenopus laevis EF1 -alpha promoter. SEQ ID NO: 6 Core Xenopus laevis EF1 -alpha promoter with tetracycline operator sites. SEQ ID NO 7 Human CMV promoter containing intron A.
SEQ ID NO 8 Simian CMV IE94 gene promoter.
SEQ ID NO 9 Human CMV immediate-early promoter.
SEQ ID NO 10 METRAP antigen.
SEQ ID NO 1 1 Enhanced green fluorescent protein (EGFP).
SEQ ID NO 12 Bovine growth hormone polyadenylation signal
SEQ ID NO 13 pSportCMV6 (Invitrogen).
SEQ ID NO 14 pKan.
SEQ ID NO 15 Photinus Luciferase.
SEQ ID NO 16 Synthetic intron from pCI (Promega).
SEQ ID NO 17 Rabbit beta-globin intron.
SEQ ID NO 18 Human EF1 -alpha promoter.
SEQ ID NO 19 Human EF1 -alpha intron.
SEQ ID NO: 1 Core EF1 -alpha promoter sequence from Xenopus laevis. g acc gagc cgCTAGCTTATGACGCACTAGGGAGTGCCACCCTTCCTTTCGCCCTAACTTCGTGATAACTCGCGCGTTTCACTCAACA GCTGCATCCGCCCTAGTGCTACTGGGAGTTGTAGTATACAAGACGCTTACAGGCTGAATGTTCTGTCAAGACCCCGCCTCTAGCACTTTG GGAATTCTGGACTTGATGATGTCATGGTTAATCCCCGCCCAGTAGAGGCGGCTATATAAAGGGTGGTTAAGGCCCGGTTCGCTCTCTTCC TCACCGGGTCTGCGGCGAGCTCTCTTAAGGTAGCGGATCCACTAGTTCTAGAAgatctggcctcggcggccaagcttggcaatccggtac tgttggtaaagccac
SEQ ID NO: 2 Enhancer sequence from SV40.
Cgatggagcggagaatgggcggaactgggcggagttaggggcgggatgggcggagttaggggcgggactatggttgctgactaattgagatgcatgctttgcatacttct gcctgctggggagcctggggactttccacacctggttgctgactaattgagatgcatgctttgcatacttctgcctgctggggagcctggggactttccacaccctaactgacac acattccacagc
SEQ ID NO: 3 Enhancer sequence from EF1 -alpha gene of Xenopus laevis.
ATCGATGGATCATCTAATCAAGCACAAATAAGGGGCGTGTAACACAAAAGCCAGCGACCCTTTCCAATGCAAATCAAACTTGCAATTCTT TGCCGTTTTTATCATTTAAGTGTCGGCTTAAGGTCCACTATCAGATGTAAACAGCCTTATCTAACAA AGGTATCATTACATTCTGAAATTCTCAGGCATGCAAG
SEQ ID NO: 4 Tetracycline operator site sequence.
TCCCTATCAGTGATAGAGATCTCCCTATCAGTGATAGAGA SEQ ID NO: 5 Full length sequence of Xenopus laevis EF1 -alpha promoter.
GTACCATCGATGGATCATCTAATCAAGCACAAATAAGGGGCGTGTAACACAAAAGCCAGCGACCCTTTCCAATGCAAATCAAACTTGCAA TTCTTTGCCGTTTTTATCATTTAAGTGTCGGCTTAAGGTCCACTATCAGATGTAAACAGCCTTATCT AACAAAGGTATCATTACATTCTGAAATTCTCAGGCATGCAAGCTAGCTTATGACGCACTAGGGAGTG CCACCCTTCCTTTCGCCCTAACTTCGTGATAACTCGCGCGTTTCACTCAACAGCTGCATCCGCCCTA GTGCTACTGGGAGTTGTAGTATACAAGACGCTTACAGGCTGAATGTTCTGTCAAGACCCCGCCTCTA GCACTTTGGGAATTCTGGACTTGATGATGTCATGGTTAATCCCCGCCCAGTAGAGGCGGCTATATAA AGGGTGGTTAAGGCCCGGTTCGCTCTCTTCCTCACCGGGTCTGCGGCGAGCTCTCTTAAGGTAGCGG ATCCACTAGTTCTAGAAgatct
SEQ ID NO: 6 Core Xenopus laevis EF1 -alpha promoter with tetracycline operator sites.
CTAGCTTATGACGCACTAGGGAGTGCCACCCTTCCTTTCGCCCTAACTTCGTGATAACTCGCGCGTTTCACTCAACAG CTGCATCCGCCCTAGTGCTACTGGGAGTTGTAGTATACAAGACGCTTACAGGCTGAAT GTTCTGTCAAGACCCCGCCTCTAGCACTTTGGGAATTCTGGACTTGATGATGTCATGG TTAATCCCCGCCCAGTAGAGGCGGCTATATAAAGGGTGGTCTCCCTATCAGTGATAGA GATCTCCCTATCAGTGATAGAGATCGTCGACGACGAGCTGGGTGGTTAAGGCCCGGT TCGCTCTCTTCCTCACCGGGTCTGCGGCGAGG
SEQ ID NO: 7 Human CMV promoter containing intron A.
TGTGAGTTTCTGTGTAACTGATATCGCCATTTTTCCAAAAGTGATTTTTGGGCATACGCGATATCTGGCGATAGCGCTT ATATCGTTTACGGGGGATGGCGATAGACGACTTTGGTGACTTGGGCGATTCTGTGTGT CGCAAATATCGCAGTTTCGATATAGGTGACAGACGATATGAGGCTATATCGCCGATAG AGGCGACATCAAGCTGGCACATGGCCAATGCATATCGATCTATACATTGAATCAATATT GGCCATTAGCCATATTATTCATTGGTTATATAGCATAAATCAATATTGGCTATTGGCCAT TGCATACGTTGTATCCATATCATAATATGTACATTTATATTGGCTCATGTCCAACATTAC CGCCATGTTGACATTGATTATTGACTAGTTATTAATAGTAATCAATTACGGGGTCATTA GTTCATAGCCCATATATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTG GCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGT AACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCC CACTTGGCAGTACATCAAGTGTATCATATGCCAAGTACGCCCCCTATTGACGTCAATG ACGGTAAATGGCCCGCCTGGCATTATGCCCAGTACATGACCTTATGGGACTTTCCTAC TTGGCAGTACATCTACGTATTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGT ACATCAATGGGCGTGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCAT TGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCAAAATGTCGT AACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGAGGTCTAT ATAAGCAGAGCTCGTTTAGTGAACCGTCAGATCGCCTGGAGACGCCATCCACGCTGTT TTGACCTCCATAGAAGACACCGGGACCGATCCAGCCTCCGCGGCCGGGAACGGTGC ATTGGAACGCGGATTCCCCGTGCCAAGAGTGACGTAAGTACCGCCTATAGAGTCTATA GGCCCACCCCCTTGGCTTCTTATGCATGCTATACTGTTTTTGGCTTGGGGTCTATACA CCCCCGCTTCCTCATGTTATAGGTGATGGTATAGCTTAGCCTATAGGTGTGGGTTATT GACCATTATTGACCACTCCCCTATTGGTGACGATACTTTCCATTACTAATCCATAACAT GGCTCTTTGCCACAACTCTCTTTATTGGCTATATGCCAATACACTGTCCTTCAGAGACT
GACACGGACTCTGTATTTTTACAGGATGGGGTCTCATTTATTATTTACAAATTCACATAT
ACAACACCACCGTCCCCAGTGCCCGCAGTTTTTATTAAACATAACGTGGGATCTCCAC
GCGAATCTCGGGTACGTGTTCCGGACATGGGCTCTTCTCCGGTAGCGGCGGAGCTTC
TACATCCGAGCCCTGCTCCCATGCCTCCAGCGACTCATGGTCGCTCGGCAGCTCCTT
GCTCCTAACAGTGGAGGCCAGACTTAGGCACAGCACGATGCCCACCACCACCAGTGT
GCCGCACAAGGCCGTGGCGGTAGGGTATGTGTCTGAAAATGAGCTCGGGGAGCGGG
CTTGCACCGCTGACGCATTTGGAAGACTTAAGGCAGCGGCAGAAGAAGATGCAGGCA
GCTGAGTTGTTGTGTTCTGATAAGAGTCAGAGGTAACTCCCGTTGCGGTGCTGTTAAC
GGTGGAGGGCAGTGTAGTCTGAGCAGTACTCGTTGCTGCCGCGCGCGCCACCAGAC
ATAATAGCTGACAGACTAACAGACTGTTCCTTTCCATGGGTCTTTTCTGCA
SEQ ID NO: 8 Simian CMV IE94 gene promoter.
GTACCATCGATGGCCGCATTGAATTGGCATGGTGCCAATAATGGCGGCCATATTGGCTATATGCCAGGATCAATATATAGGCAATATCCA ATATGGCCCTATGCCAATATGGCTATTGGCCAGGTTCAATACTATGTATTGGCCCTATGCCATATAG TATTCCATATATGGGTTTTCCTATTGACGTAGATAGCCCCTCCCAATGGGCGGTCCCATATACCATA TATGGGGCTTCCTAATACCGCCCATAGCCACTCCCCCATTGACGTCAATGGTCTCTATATATGGTCT TTCCTATTGACGTCATATGGGCGGTCCTATTGACGTATATGGCGCCTCCCCCATTGACGTCAATTAC GGTAAATGGCCCGCCTGGCTCAATGCCCATTGACGTCAATAGGACCACCCACCATTGACGTCAATGG GATGGCTCATTGCCCATTCATATCCGTTCTCACGCCCCCTATTGACGTCAATGACGGTAAATGGCCC ACTTGGCAGTACATCAATATCTATTAATAGTAACTTGGCAAGTACATTACTATTGGCAAGTACGCCA AGGGTACATTGGCAGGTACTCCCATTGACGTCAATGGCGGTAAATGGCCCGGCATGGCTGCCAAGTA CAACATCCCCATTGACGTCAATGGGAAGGGGCAATGACGCAAATGGGCGTTCCATTGACGTAAATGG CGGTAGGCGTGCCTAATGGGAGGTCTATATAAGCAATGCTCGTTTAGGGAACCGCCATTCTGCCTGG GGACGTCGGAGGAGCTCCATTGGAAGAGACCGGGACCGATCCAGCCTCCATAGCCGGGGACGGTGCA TTGGAATGCGGATAGAGCTTGCCAAG
SEQ ID NO: 9 Human CMV immediate-early promoter.
CGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGT ATGTTCCCATAGTAACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTA CGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCATATGCCAAGTACGCCCCCTA TTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGCCCAGTACATGACCTTATG GGACTTTCCTACTTGGCAGTACATCTACGTATTAGTCATCGCTATTACCATGGTGATGC GGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCACGGGGATTTCCAA GTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTT TCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTACG GTGGGAGGTCTATATAAGCAGAGCTCGTTTAGTGAACCGTCAGATCGCCTGGAGACG CCATCCACGCTGTTTTGACCTCCATAGAAGACACCGGGACCGATCCAGCCT
SEQ ID NO: 10 METRAP antigen.
ATGATCAACGCCTACTTGGACAAGTTGATCTCCAAGTACGAAGACGAAATCTCCTACATCCCATCTGCCGAAAAGATC GGATCTAAGCCGAACGACAAGTCCTTGTATAAACCTAAGGACGAATTGGACTACAAGC CAATCGTTCAATACGACAACTTCGGATCTGCCTCCAAGAACAAGGAAAAGGCTTTGAT
CATCGGTATCGCTGGTGGTTTGGCCTTGTTGATGAACCCTAATGACCCAAACAGAAAC
GTCAGATCTCACTTGGGTAACGTTAAGTACTTGGTTAAGTCTTTGTACGATGAACACAT
CTTATTGATGGACTGTTCTGGTTCTATTGGATCTGACCCAAACGCTAACCCAAACGTTG
ACCCAAACGCCAACCCAAACGTCCAAGTTCACTTCCAACCATTGCCTCCGGCCGTTGT
CAAGTTGCAATTCATCAAGGCCAACTCTAAGTTCATCGGTATCACCGAAGGATCTTACT
TGAACAAAATTCAAAACTCTTTGATGGAAAAGTTGAAAGAATTGGAAAAGGCTACTTCT
GTCTTGGCTGGTTTGGGATCTAACGCTAATCCAAACGCAAATCCGAACGCCAATCCTA
ACGCGAATCCCGACGAATGGTCTCCATGTTCTGTCACTTGTGGTAAGGGTACTCGCTC
TAGAAAGAGAGAAGGATCCAAAATAATGAATCATCTTGGGAATGTTAAATATTTAGTCA
TTGTGTTTTTGATTTTCTTTGATTTGTTTCTAGTTAATGGTAGAGATGTGCAAAACAATA
TAGTGGATGAAATAAAATATAGTGAAGAAGTATGTAATGATCAGGTAGATCTTTACCTT
CTAATGGATTGTTCTGGAAGTATACGTCGTCATAATTGGGTGAACCATGCAGTACCTCT
AGCTATGAAATTGATACAACAATTAAATCTTAATGATAATGCAATTCACTTATATGTTAAT
GTTTTTTCAAACAATGCAAAAGAAATTATTAGATTACATAGTGATGCATCTAAAAACAAA
GAGAAGGCTTTAATTATTATAAGGTCACTCTTAAGTACAAATCTTCCATATGGTAGAAC
AAACTTAACTGATGCACTGTTACAAGTAAGAAAACATTTAAATGACCGAATCAATAGAG
AGAATGCTAATCAATTAGTTGTTATATTAACAGATGGAATTCCAGATAGTATTCAAGATT
CATTAAAAGAATCAAGAAAATTAAGTGATCGTGGTGTTAAAATAGCTGTTTTTGGTATTG
GACAAGGTATTAATGTAGCTTTCAACAGATTTCTTGTAGGTTGTCATCCATCAGATGGT
AAATGTAACTTGTATGCTGATTCTGCATGGGAAAATGTAAAAAATGTTATCGGACCCTT
TATGAAGGCTGTTTGTGTTGAAGTAGAAAAAACAGCAAGTTGTGGTGTTTGGGACGAA
TGGTCTCCATGTAGTGTAACTTGTGGTAAAGGTACCAGGTCAAGAAAAAGAGAAATCT
TACACGAAGGATGTACAAGTGAAATACAAGAACAATGTGAAGAAGAAAGATGTCCTCC
AAAATGGGAACCATTAGATGTTCCAGATGAACCCGAAGATGATCAACCTAGACCAAGA
GGAGATAATTCTTCTGTCCAAAAACCAGAAGAAAATATAATAGATAATAATCCACAAGA
ACCTTCACCAAATCCAGAAGAAGGAAAGGATGAAAATCCAAACGGATTTGATTTAGAT
GAAAATCCAGAAAATCCACCAAATCCAGATATTCCTGAACAAAAACCAAATATACCTGA
AGATTCAGAAAAAGAAGTACCTTCTGATGTTCCAAAAAATCCAGAAGACGATCGAGAA
GAAAACTTTGATATTCCAAAGAAACCCGAAAATAAGCACGATAATCAAAATAATTTACC
AAATGATAAAAGTGATAGAAATATTCCATATTCACCATTACCTCCAAAAGTTTTGGATAA
TGAAAGGAAACAAAGTGACCCCCAAAGTCAAGATAATAATGGAAATAGGCACGTACCT
AATAGTGAAGATAGAGAAACACGTCCACATGGTAGAAATAATGAAAATAGATCATACAA
TAGAAAATATAACGATACTCCAAAACATCCTGAAAGGGAAGAACATGAAAAGCCAGATA
ATAATAAAAAAAAAGGAGAATCAGATAATAAATATAAAATTGCAGGTGGAATAGCTGGA
GGATTAGCTTTACTCGCATGTGCTGGACTTGCTTATAAATTCGTAGTACCAGGAGCAG
CAACACCCTATGCCGGAGAACCTGCACCTTTTGATGAAACATTAGGTGAAGAAGATAA
AGATTTGGACGAACCTGAACAATTCAGATTACCTGAAGAAAACGAGTGGAATTAAA
SEQ ID NO: 1 1 Enhanced green fluorescent protein (EGFP).
ATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCGAGCTGGACGGCGACGTAAACGGC CACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGCGATGCCACCTACGGCAAGCTGAC CCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTGCCCTGGCCCACCCTCGTGAC CACCCTGACCTACGGCGTGCAGTGCTTCAGCCGCTACCCCGACCACATGAAGCAGCA CGACTTCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTCTTC AAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACCCT
GGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAACATCCTGG
GGCACAAGCTGGAGTACAACTACAACAGCCACAACGTCTATATCATGGCCGACAAGCA
GAAGAACGGCATCAAGGTGAACTTCAAGATCCGCCACAACATCGAGGACGGCAGCGT
GCAGCTCGCCGACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGC
TGCCCGACAACCACTACCTGAGCACCCAGTCCGCCCTGAGCAAAGACCCCAACGAGA
AGCGCGATCACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCA
TGGACGAGCTGTACAAGTAA
SEQ ID NO: 12 Bovine growth hormone polyadenylation signal.
TAGAGGGCCCTATTCTATAGTGTCACCTAAATGCTAGAGCTCGCTGATCAGCCTCGACTGTGCCTTCTAGTTGCCAGC CATCTGTTGTTTGCCCCTCCCCCGTGCCTTCCTTGACCCTGGAAGGTGCCACTCCCAC TGTCCTTTCCTAATAAAATGAGGAAATTGCATCGCATTGTCTGAGTAGGTGTCATTCTA TTCTGGGGGGTGGGGTGGGGCAGGACAGCAAGGGGGAGGATTGGGAAGACAATAGC AGGCATGCTGGGGATGCGGTGGGCTCTATGGCTTCTGAGGCGGAAAGAACCAGCTG GGGC
SEQ ID NO: 13 pSportCMV6 (Invitrogen).
CCATTCGCCA TTCAGGCTGC GCAACTGTTG GGAAGGGCGA TCGGTGCGGG CCTCTTCGCT 60
ATTACGCCAG CCAATACGCA AACCGCCTCT CCCCGCGCGT TGGCCGATTC ATTAATGCAG 120
GATCGATCCA GACATGATAA GATACATTGA TGAGTTTGGA CAAACCACAA CTAGAATGCA 180
GTGAAAAAAA TGCTTTATTT GTGAAATTTG TGATGCTATT GCTTTATTTG TAACCATTAT 240
AAGCTGCAAT AAACAAGTTA ACAACAACAA TTGCATTCAT TTTATGTTTC AGGTTCAGGG 300
GGAGGTGTGG GAGGTTTTTT AAAGCAAGTA AAACCTCTAC AAATGTGGTA TGGCTGATTA 360
TGATCATGAA CAGACTGTGA GGACTGAGGG GCCTGAAATG AGCCTTGGGA CTGTGAATCT 420
AAAATACACA AACAATTAGA ATCACTAGCT CCTGTGTATA ATATTTTCAT AAATCATACT 480
CAGTAAGCAA AACTCTCAAG CAGCAAGCAT ATGCAGCTAG TTTAACACAT TATACACTTA 540
AAAATTTTAT ATTTACCTTA GAGCTTTAAA TCTCTGTAGG TAGTTTGTCC AATTATGTCA 600
CACCACAGAA GTAAGGTTCC TTCACAAAGA TCCCAAGCTA GCAGTTTTCC CAGTCACGAC 660
GTTGTAAAAC GACGGCCAGT GCCTAGCTTA TAATACGACT CACTATAGGG ACCACTTTGT 720
ACAAGAAAGC TGGGTACGCG TAAGCTTGGG CCCCTCGAGG GATACTCTAG AGCGGCCgcg 780 aattcagatc tcacATGtTG GCGGCaccgg tggatccggt accAGCCTGC TTTTTTGTAC 840
AAACTTGTTC TATAGTGTCA CCTAAATAGG CCTAATGGTC ATAGCTGTTT CCTGTGTGAA 900
ATTGTTATCC GCTCCGCGGC CTAGGCTAGA GTCCGGAGGC TGGATCGGTC CCGGTGTCTT 960
CTATGGAGGT CAAAACAGCG TGGATGGCGT CTCCAGGCGA TCTGACGGTT CACTAAACGA 1020
GCTCTGCTTA TATAGACCTC CCACCGTACA CGCCTACCGC CCATTTGCGT CAATGGGGCG 1080
GAGTTGTTAC GACATTTTGG AAAGTCCCGT TGATTTTGGT GCCAAAACAA ACTCCCATTG 1140
ACGTCAATGG GGTGGAGACT TGGAAATCCC CGTGAGTCAA ACCGCTATCC ACGCCCATTG 1200
ATGTACTGCC AAAACCGCAT CACCATGGTA ATAGCGATGA CTAATACGTA GATGTACTGC 1260
CAAGTAGGAA AGTCCCATAA GGTCATGTAC TGGGCATAAT GCCAGGCGGG CCATTTACCG 1320
TCATTGACGT CAATAGGGGG CGTACTTGGC ATATGATACA CTTGATGTAC TGCCAAGTGG 1380
GCAGTTTACC GTAAATACTC CACCCATTGA CGTCAATGGA AAGTCCCTAT TGGCGTTACT 1440
ATGGGAACAT ACGTCATTAT TGACGTCAAT GGGCGGGGGT CGTTGGGCGG TCAGCCAGGC 1500
GGGCCATTTA CCGTAAGTTA TGTAACGACA TGCATCTAAT GAGTGAAAGG GCCTCGTACT 1560
ACGCCTATTT TTATAGGTTA ATGTCATGAT AATAATGGTT TCTTAGACGT CAGGTGGCAC 1620
TTTTCGGGGA AATGTGCGCG GAACCCCTAT TTGTTTATTT TTCTAAATAC ATTCAAATAT 1680
GTATCCGCTC ATGAGACAAT AACCCTGATA AATGCTTCAA TAATATTGAA AAACGCGCGA 1740
ATTGCAAGCT CTGCATTAAT GAATCGGCCA ACGCGCGGGG AGAGGCGGTT TGCGTATTGG 1800
GCGCTCTTCC GCTTCCTCGC TCACTGACTC GCTGCGCTCG GTCGTTCGGC TGCGGCGAGC 1860
GGTATCAGCT CACTCAAAGG CGGTAATACG GTTATCCACA GAATCAGGGG ATAACGCAGG 1920
AAAGAAcatC ATGTGAGCAA AAGGCCAGCA AAAGGCCAGG AACCGTAAAA AGGCCGCGTT 1980
GCTGGCGTTT TTCCATAGGC TCCGCCCCCC TGACGAGCAT CACAAAAATC GACGCTCAAG 2040
TCAGAGGTGG CGAAACCCGA CAGGACTATA AAGATACCAG GCGTTTCCCC CTGGAAGCTC 2100
CCTCGTGCGC TCTCCTGTTC CGACCCTGCC GCTTACCGGA TACCTGTCCG CCTTTCTCCC 2160
TTCGGGAAGC GTGGCGCTTT CTCAATGCTC ACGCTGTAGG TATCTCAGTT CGGTGTAGGT 2220
CGTTCGCTCC AAGCTGGGCT GTGTGCACGA ACCCCCCGTT CAGCCCGACC GCTGCGCCTT 2280
ATCCGGTAAC TATCGTCTTG AGTCCAACCC GGTAAGACAC GACTTATCGC CACTGGCAGC 2340
AGCCACTGGT AACAGGATTA GCAGAGCGAG GTATGTAGGC GGTGCTACAG AGTTCTTGAA 2400 GTGGTGGCCT AACTACGGCT ACACTAGAAG GACAGTATTT GGTATCTGCG CTCTGCTGAA 2460
GCCAGTTACC TTCGGAAAAA GAGTTGGTAG CTCTTGATCC GGCAAACAAA CCACCGCTGG 2520
TAGCGGTGGT TTTTTTGTTT GCAAGCAGCA GATTACGCGC AGAAAAAAAG GATCTCAAGA 2580
AGATCCTTTG ATCTTTTCTA CGGGGTCTGA CGCTCAGTGG AACGAAAACT CACGTTAAGG 2640
GATTTTGGTC ATGAGATTAT CAAAAAGGAT CTTCACCTAG ATCCTTTTAA ATTAAAAATG 2700
AAGTTTTAAA TCAATCTAAA GTATATATGA GTAAACTTGG TCTGACAGTT ACCAATGCTT 2760
AATCAGTGAG GCACCTATCT CAGCGATCTG TCTATTTCGT TCATCCATAG TTGCCTGACT 2820
CCCCGTCGTG TAGATAACTA CGATACGGGA GGGCTTACCA TCTGGCCCCA GTGCTGCAAT 2880
GATACCGCGA GACCCACGCT CACCGGCTCC AGATTTATCA GCAATAAACC AGCCAGCCGG 2940
AAGGGCCGAG CGCAGAAGTG GTCCTGCAAC TTTATCCGCC TCCATCCAGT CTATTAATTG 3000
TTGCCGGGAA GCTAGAGTAA GTAGTTCGCC AGTTAATAGT TTGCGCAACG TTGTTGCCAT 3060
TGCTACAGGC ATCGTGGTGT CACGCTCGTC GTTTGGTATG GCTTCATTCA GCTCCGGTTC 3120
CCAACGATCA AGGCGAGTTA CATGATCCCC CATGTTGTGC AAAAAAGCGG TTAGCTCCTT 3180
CGGTCCTCCG ATCGTTGTCA GAAGTAAGTT GGCCGCAGTG TTATCACTCA TGGTTATGGC 3240
AGCACTGCAT AATTCTCTTA CTGTCATGCC ATCCGTAAGA TGCTTTTCTG TGACTGGTGA 3300
GTACTCAACC AAGTCATTCT GAGAATAGTG TATGCGGCGA CCGAGTTGCT CTTGCCCGGC 3360
GTCAATACGG GATAATACCG CGCCACATAG CAGAACTTTA AAAGTGCTCA TCATTGGAAA 3420
ACGTTCTTCG GGGCGAAAAC TCTCAAGGAT CTTACCGCTG TTGAGATCCA GTTCGATGTA 3480
ACCCACTCGT GCACCCAACT GATCTTCAGC ATCTTTTACT TTCACCAGCG TTTCTGGGTG 3540
AGCAAAAACA GGAAGGCAAA ATGCCGCAAA AAAGGGAATA AGGGCGACAC GGAAATGTTG 3600
AATACTCATA CTCTTCCTTT TTCAATATTA TTGAAGCATT TATCAGGGTT ATTGTCTCAT 3660
GAGCGGATAC ATATTTGAAT GTATTTAGAA AAATAAACAA ATAGGGGTTC CGCGCACATT 3720
TCCCCGAAAA GTGCCACCTG AAATTGTAAA CGTTAATATT TTGTTAAAAT TCGCGTTAAA 3780
TTTTTGTTAA ATCAGCTCAT TTTTTAACCA ATAGGCCGAA ATCGGCAAAA TCCCTTATAA 3840
ATCAAAAGAA TAGACCGAGA TAGGGTTGAG TGTTGTTCCA GTTTGGAACA AGAGTCCACT 3900
ATTAAAGAAC GTGGACTCCA ACGTCAAAGG GCGAAAAACC GTCTATCAGG GCGATGGCCC 3960
ACTACGTGAA CCATCACCCT AATCAAGTTT TTTGGGGTCG AGGTGCCGTA AAGCACTAAA 4020
TCGGAACCCT AAAGGGAGCC CCCGATTTAG AGCTTGACGG GGAAAGCCGG CGAACGTGGC 4080
GAGAAAGGAA GGGAAGAAAG CGAAAGGAGC GGGCGCTAGG GCGCTGGCAA GTGTAGCGGT 4140
CACGCTGCGC GTAACCACCA CACCCGCCGC GCTTAATGCG CCGCTACAGG GCGCGTC 4200
SEQ ID NO: 14 pKan.
1 CTTTCCTGCG TTATCCCCTG ATTCTGTGGA TAACCGTATT ACCGCTAGCA TGGATCTCGG
61 GGACGTCTAA CTACTAAGCG AGAGTAGGGA ACTGCCAGGC ATCAAATAAA ACGAAAGGCT
121 CAGTCGGAAG ACTGGGCCTT TCGTTTTATC TGTTGTTTGT CGGTGAACGC TCTCCTGAGT
181 AGGACAAATC CGCCGGGAGC GGATTTGAAC GTTGTGAAGC AACGGCCCGG AGGGTGGCGG
241 GCAGGACGCC CGCCATAAAC TGCCAGGCAT CAAACTAAGC AGAAGGCCAT CCTGACGGAT
301 GGCCTTTTTG CGTTTCTACA AACTCTTCCT GTTAGTTAGT TACTTAAGCT CGGGCCCCAA
361 ATAATGATTT TATTTTGACT GATAGTGACC TGTTCGTTGC AACAAATTGA TAAGCAATGC
421 TTTTTTATAA TGCCAACTTT GTACAAAAAA GCAGGCTCCA CCATGGGAAC CAATTCAGTC
481 gactctcagt acaatctgct ctgatgccgc atagttaagc cagtatctgc tccctgcttg
541 tgtgttggag gtcgctgagt agtgcgcgag caaaatttaa gctacaacaa ggcaaggctt
601 gaccgacaat tgcatgaaga atctgcttag ggttaggcgt tttgcgctgc ttcgcgatgt
661 acgggccaga tataCGCGCA ATTAACCCTC ACTAAAGGGA ACAAAAGCTG GAGCTCGGCC
721 CTCTAGGCCC CTAGGCCATA ACATTCTGAC ACACTCCCGC CCAGGAGGGG AGACGCCCCG
781 TCAGATAAGG GCTGAATACC CCTCCCTCAA AGGAGGCGCT TCCGCTCATG CTGCTGCAAC
841 ACACAGAAAA CCACAGATTT TAGTCATTCT GCCCCTCATG CAGGCTCACA CCCAGCCCCA
901 AACACTACTC ACGCGATCGC TCGGTCGAcg gatccgctgt ggaatgtgtg tcagttaggg
961 tgtggaaagt ccccaggctc cccagcaggc agaagtatgc aaagcatgca tctcaattag
1021 tcagcaacca ggtgtggaaa gtccccaggc tccccagcag gcagaagtat gcaaagcatg
1081 catctcaatt agtcagcaac catagtcccg cccctaactc cgcccatccc gcccctaact
1141 ccgcccagtt ccgcccattc tccgctccat cgttcagatc cttatcgatt ttaccacatt
1201 tgtagaggtt ttacttgctt taaaaaacct cccacacctc cccctgaacc tgaaacataa
1261 aatgaatgca attgttgttg ttaacttgtt tattgcagct tataatggtt acaaataaag
1321 caatagcatc acaaatttca caaataaagc atttttttca ctgcattcta gttgtggttt
1381 gtccaaactc atcaatgtat cttatcatgt ctgctcgaag cggccggccG CACGCGTCCC
1441 GGGAGATCTG GTACCGAATT CGCTAGCAAG AGCTAGGATC CATCGATCTC GAGCTAGAGC
1501 CCACCGCATC CCCAGCATGC CTGCTATTGT CTTCCCAATC CTCCCCCTTG CTGTCCTGCC
1561 CCACCCCACC CCCCAGAATA GAATGACACC TACTCAGACA ATGCGATGCA ATTTCCTCAT
1621 TTTATTAGGA AAGGACAGTG GGAGTGGCAC CTTCCAGGGT CAAGGAAGGC ACGGCCCTCT
1681 AGGCCTCTAG ACCCAGCTTT CTTGTACAAA GTTGGCATTA TAAGAAAGCA TTGCTTATCA
1741 ATTTGTTGCA ACGAACAGGT CACTATCAGT CAAAATAAAA TCATTATTTG CCATCCAGCT 1801 GCAGCTCTGG CCCGTGTCTC AAAATCTCTG ATGTTACATT GCACAAGATA AAAATATATC
1861 ATCATGAACA ATAAAACTGT CTGCTTACAT AAACAGTAAT ACAAGGGGTG TTATGAGCCA 1921 TATTCAACGG GAAACGTCGA GGCCGCGATT AAATTCCAAC ATGGATGCTG ATTTATATGG
1981 GTATAAATGG GCTCGCGATA ATGTCGGGCA ATCAGGTGCG ACAATCTATC GCTTGTATGG 2041 GAAGCCCGAT GCGCCAGAGT TGTTTCTGAA ACATGGCAAA GGTAGCGTTG CCAATGATGT 2101 TACAGATGAG ATGGTCAGAC TAAACTGGCT GACGGAATTT ATGCCTCTTC CGACCATCAA
2161 GCATTTTATC CGTACTCCTG GTGATGCATG GTTACTCACC ACTGCGATCC CCGGAAAAAC
2221 AGCATTCCAG GTATTAGAAG AATATCCTGA TTCAGGTGAA AATATTGTTG ATGCGCTGGC
2281 AGTGTTCCTG CGCCGGTTGC ATTCGATTCC TGTTTGTAAT TGTCCTTTTA ACAGCGATCG
2341 CGTATTTCGT CTCGCTCAGG CGCAATCACG AATGAATAAC GGTTTGGTTG ATGCGAGTGA
2401 TTTTGATGAC GAGCGTAATG GCTGGCCTGT TGAACAAGTC TGGAAAGAAA TGCATAAACT
2461 TTTGCCATTC TCACCGGATT CAGTCGTCAC TCATGGTGAT TTCTCACTTG ATAACCTTAT
2521 TTTTGACGAG GGGAAATTAA TAGGTTGTAT TGATGTTGGA CGAGTCGGAA TCGCAGACCG
2581 ATACCAGGAT CTTGCCATCC TATGGAACTG CCTCGGTGAG TTTTCTCCTT CATTACAGAA
2641 ACGGCTTTTT CAAAAATATG GTATTGATAA TCCTGATATG AATAAATTGC AGTTTCATTT
2701 GATGCTCGAT GAGTTTTTCT AATCAGAATT GGTTAATTGG TTGTAACATT ATTCAGATTG
2761 GGCCCCGTTC CACTGAGCGT CAGACCCCGT AGAAAAGATC AAAGGATCTT CTTGAGATCC
2821 TTTTTTTCTG CGCGTAATCT GCTGCTTGCA AACAAAAAAA CCACCGCTAC CAGCGGTGGT
2881 TTGTTTGCCG GATCAAGAGC TACCAACTCT TTTTCCGAAG GTAACTGGCT TCAGCAGAGC
2941 GCAGATACCA AATACTGTTC TTCTAGTGTA GCCGTAGTTA GGCCACCACT TCAAGAACTC
3001 TGTAGCACCG CCTACATACC TCGCTCTGCT AATCCTGTTA CCAGTGGCTG CTGCCAGTGG
3061 CGATAAGTCG TGTCTTACCG GGTTGGACTC AAGACGATAG TTACCGGATA AGGCGCAGCG
3121 GTCGGGCTGA ACGGGGGGTT CGTGCACACA GCCCAGCTTG GAGCGAACGA CCTACACCGA
3181 ACTGAGATAC CTACAGCGTG AGCTATGAGA AAGCGCCACG CTTCCCGAAG GGAGAAAGGC
3241 GGACAGGTAT CCGGTAAGCG GCAGGGTCGG AACAGGAGAG CGCACGAGGG AGCTTCCAGG
3301 GGGAAACGCC TGGTATCTTT ATAGTCCTGT CGGGTTTCGC CACCTCTGAC TTGAGCGTCG
3361 ATTTTTGTGA TGCTCGTCAG GGGGGCGGAG CCTATGGAAA AACGCCAGCA ACGCGGCCTT
3421 TTTACGGTTC CTGGCCTTTT GCTGGCCTTT TGCTCACATG TT
SEQ ID NO: 15 Photinus Luciferase.
atggaagatgccaaaaacattaagaagggcccagcgccattctacccactcgaagacgggaccgccggcgagcagctgcacaaagccatgaagcgctacgccctggt gcccggcaccatcgcctttaccgacgcacatatcgaggtggacattacctacgccgagtacttcgagatgagcgttcggctggcagaagctatgaagcgctatgggctgaa tacaaaccatcggatcgtggtgtgcagcgagaatagcttgcagttcttcatgcccgtgttgggtgccctgttcatcggtgtggctgtggccccagctaacgacatctacaacga gcgcgagctgctgaacagcatgggcatcagccagcccaccgtcgtattcgtgagcaagaaagggctgcaaaagatcctcaacgtgcaaaagaagctaccgatcataca aaagatcatcatcatggatagcaagaccgactaccagggcttccaaagcatgtacaccttcgtgacttcccatttgccacccggcttcaacgagtacgacttcgtgcccgag agcttcgaccgggacaaaaccatcgccctgatcatgaacagtagtggcagtaccggattgcccaagggcgtagccctaccgcaccgcaccgcttgtgtccgattcagtcat gcccgcgaccccatcttcggcaaccagatcatccccgacaccgctatcctcagcgtggtgccatttcaccacggcttcggcatgttcaccacgctgggctacttgatctgcgg ctttcgggtcgtgctcatgtaccgcttcgaggaggagctattcttgcgcagcttgcaagactataagattcaatctgccctgctggtgcccacactatttagcttcttcgctaagag cactctcatcgacaagtacgacctaagcaacttgcacgagatcgccagcggcggggcgccgctcagcaaggaggtaggtgaggccgtggccaaacgcttccacctacc aggcatccgccagggctacggcctgacagaaacaaccagcgccattctgatcacccccgaaggggacgacaagcctggcgcagtaggcaaggtggtgcccttcttcga ggctaaggtggtggacttggacaccggtaagacactgggtgtgaaccagcgcggcgagctgtgcgtccgtggccccatgatcatgagcggctacgttaacaaccccgag gctacaaacgctctcatcgacaaggacggctggctgcacagcggcgacatcgcctactgggacgaggacgagcacttcttcatcgtggaccggctgaagagcctgatca aatacaagggctaccaggtagccccagccgaactggagagcatcctgctgcaacaccccaacatcttcgacgccggggtcgccggcctgcccgacgacgatgccggc gagctgcccgccgcagtcgtcgtgctggaacacggtaaaaccatgaccgagaaggagatcgtggactatgtggccagccaggttacaaccgccaagaagctgcgcggt ggtgttgtgttcgtggacgaggtgcctaaaggactgaccggcaagttggacgcccgcaagatccgcgagattctcattaaggccaagaagggcggcaagatcgccgtgta
SEQ ID NO: 16 Synthetic intron from pCI (Promega).
gtaagtatcaaggttacaagacaggtttaaggagaccaatagaaactgggcttgtcgagacagagaagactcttgcgtttctgataggcacctattggtcttactgacatcca ctttgcctttctctccacag
SEQ ID NO: 17 Rabbit beta-globin intron.
gtgagtttggggacccttgattgttctttctttttcgctattgtaaaattcatgttatatggagggggra
accctcatgataattttgtttctttcactttctactctgttgacaaccattgtctcrt^ tttgtttatttgtcagattgtaagtactttctctaatcacttttttttcaaggcaatcagggtatattatattgtacttcagcacagttttagagaacaattgttataattaa aatatttctgcatataaattctggctggcgtggaaatattcttattggtagaaacaactacatcctggtcatcatcctgcctttctctttatggttacaatgatatacactgt¾ aggataaaatactctgagtccaaaccgggcccctctgctaaccatgttcatgccttcttctttttcctacag
SEQ ID NO: 18 Human EF1 -alpha promoter.
GTGAGGCTCCGGTGCCCGTCAGTGGGCAGAGCGCACATCGCCCACAGTCCCCGAGAAGTTGGGGGGAGGGGTCG
GCAATTGAACCGGTGCCTAGAGAAGGTGGCGCGGGGTAAACTGGGAAAGTGATGTCGTGTACTGGCTCCGCCTTTT
TCCCGAGGGTGGGGGAGAACCGTATATAAGTGCAGTAGTCGCCGTGAACGTTCTTTTTCGCAACGGGTTTGCCGCC
AGAACACAGGTAAGTGCCGTGTGTGGTTCCCGCGGGCCTGGCCTCTTTACGGGTTATGGCCCTTGCGTGCCTTGAA
TTACTTCCACCTGGCTGCAGTACGTGATTCTTGATCCCGAGCTTCGGGTTGGAAGTGGGTGGGAGAGTTCGAGGCCT
TGCGCTTAAGGAGCCCCTTCGCCTCGTGCTTGAGTTGAGGCCTGGCCTGGGCGCTGGGGCCGCCGCGTGCGAATC
TGGTGGCACCTTCGCGCCTGTCTCGCTGCTTTCGATAAGTCTCTAGCCATTTAAAATTTTTGATGACCTGCTGCGACG
CTTTTTTTCTGGCAAGATAGTCTTGTAAATGCGGGCCAAGATCTGCACACTGGTATTTCGGTTTTTGGGGCCGCGGGC
GGCGACGGGGCCCGTGCGTCCCAGCGCACATGTTCGGCGAGGCGGGGCCTGCGAGCGCGGCCACCGAGAATCGG
ACGGGGGTAGTCTCAAGCTGGCCGGCCTGCTCTGGTGCCTGGCCTCGCGCCGCCGTGTATCGCCCCGCCCTGGGC
GGCAAGGCTGGCCCGGTCGGCACCAGTTGCGTGAGCGGAAAGATGGCCGCTTCCCGGCCCTGCTGCAGGGAGCTC
AAAATGGAGGACGCGGCGCTCGGGAGAGCGGGCGGGTGAGTCACCCACACAAAGGAAAAGGGCCTTTCCGTCCTC
AGCCGTCGCTTCATGTGACTCCACGGAGTACCGGGCGCCGTCCAGGCACCTCGATTAGTTCTCGAGCTTTTGGAGTA
CGTCGTCTTTAGGTTGGGGGGAGGGGTTTTATGCGATGGAGTTTCCCCACACTGAGTGGGTGGAGACTGAAGTTAG
GCCAGCTTGGCACTTGATGTAATTCTCCTTGGAATTTGCCCTTTTTGAGTTTGGATCTTGGTTCATTCTCAAGCCTCAG
ACAGTGGTTCAAAGTTTTTTTCTTCCATTTCAGGTGTCGTGAGGAATTCT
SEQ ID NO: 19 Human EF1 -alpha intron.
GTAAGTGCCGTGTGTGGTTCCCGCGGGCCTGGCCTCTTTACGGGTTATGGCCCTTGCGTGCCTTGAATTACTTCCAC
CTGGCTGCAGTACGTGATTCTTGATCCCGAGCTTCGGGTTGGAAGTGGGTGGGAGAGTTCGAGGCCTTGCGCTTAA
GGAGCCCCTTCGCCTCGTGCTTGAGTTGAGGCCTGGCCTGGGCGCTGGGGCCGCCGCGTGCGAATCTGGTGGCAC
CTTCGCGCCTGTCTCGCTGCTTTCGATAAGTCTCTAGCCATTTAAAATTTTTGATGACCTGCTGCGACGCTTTTTTTCT
GGCAAGATAGTCTTGTAAATGCGGGCCAAGATCTGCACACTGGTATTTCGGTTTTTGGGGCCGCGGGCGGCGACGG
GGCCCGTGCGTCCCAGCGCACATGTTCGGCGAGGCGGGGCCTGCGAGCGCGGCCACCGAGAATCGGACGGGGGT
AGTCTCAAGCTGGCCGGCCTGCTCTGGTGCCTGGCCTCGCGCCGCCGTGTATCGCCCCGCCCTGGGCGGCAAGGC
TGGCCCGGTCGGCACCAGTTGCGTGAGCGGAAAGATGGCCGCTTCCCGGCCCTGCTGCAGGGAGCTCAAAATGGA
GGACGCGGCGCTCGGGAGAGCGGGCGGGTGAGTCACCCACACAAAGGAAAAGGGCCTTTCCGTCCTCAGCCGTCG
CTTCATGTGACTCCACGGAGTACCGGGCGCCGTCCAGGCACCTCGATTAGTTCTCGAGCTTTTGGAGTACGTCGTCT
TTAGGTTGGGGGGAGGGGTTTTATGCGATGGAGTTTCCCCACACTGAGTGGGTGGAGACTGAAGTTAGGCCAGCTT
GGCACTTGATGTAATTCTCCTTGGAATTTGCCCTTTTTGAGTTTGGATCTTGGTTCATTCTCAAGCCTCAGACAGTGGT
TCAAAGTTTTTTTCTTCCATTTCAGGTGTCGTGAG List of figures Figure 1
A series of vectors were constructed expressing Photinus luciferase under the control of a series of promoters. Some promoters contained natural or synthetic splice signals (Table 1 ). These included the human CMV immediate early cassette 94 promoter (hCMV), the human EF1 a promoter (hEF1 ), the simian CMV immediate early 94 promoter (sCMV), and the Xenopus EF1 a promoter(Johnson and Krieg 1994; Johnson and Krieg 1995). Some constructs also contained the SV40 enhancer. A schematic organisation of the various promoters tested is shown above. Numbers before each construct are plasmid identifiers; sequences of the promoter and polyadenylation signal in each vector are shown in table 1. The activities shown are mean and standard deviation of three replicate transfections, in which 200ng of plasmid was co-transfected with 20ng of pGL4.74 (Promega, which expresses Renilla luciferase under the control of the TK promoter. pGL4 is genbank sequence AY738222), and luciferase measurement were made with a Dual Luciferase assay (Promega). Promoter arrangements used in adenoviruses are indicated *. Figure 2
A schematic indicating the modification of the parental AdC63-METRAP DNA by insertion of a tandem expression cassette.
Figure 3
Restriction digest of AdC63-sCMV-GFP (LEFT) or AdC63-xEF1 -GFP and AdC63-xEF1 -GFP-SV40 (RIGHT). Arrows show bands indicative of instability in the AdC63-sCMV-GFP.
Figure 4
Immunofluorescent detection of the products of both gene cassettes was performed following infection of 293 cells (in which the virus replicates) with limiting quantities of virus. Expression of both proteins was observed in all cells examined; representative cells are shown.
Figure 5
2 weeks after immunisation with AdC63-METRAP-xEF1 -GFP, AdC63-METRAP- xEF1 -GFP-SV40, or AdC63-METRAP, spleen ELISPOT assays were performed using peptides present in ME-TRAP or EGFP.
Figure 6
Activity analysis of xEF1 and xEF1.To promoter was performed by flow cytometry (Figure 6). Plasmids containing the promoters driving EGFP were transfected into either 293 or 293.Trex (Invitrogen). Flow cytometry shows both promoters have comparable activity to xEF1 when transfected into 293 cells, which lack the Tetracycline repressor protein. By contrast, substantial repression was seen when the same constructs were transfected into 293.tRex (Invitrogen) cells, in which the Tetracycline repressor is expressed.
Table 1
Vectors used in Examples.
Vector Backbone Promoter Intron Marker Polyadenylation code gene signal/enhancer
1734 Derivative of Human Nil Photinus Bovine growth pSportCMV6 CMV Luciferase hormone (Invitrogen)
1590 pGL4 Simian Nil Photinus SV40 poly A
(Promega) CMV IE94 Luciferase
1758 pGL4 Core Nil Photinus SV40 poly A &
(Promega) xEF1 a Luciferase enhancer
1752 pGL4 Complete Synthetic, Photinus SV40 poly A &
(Promega) xEF1 a from pCI Luciferase enhancer
(Promega)
1740 pGL4 Core Synthetic, Photinus SV40 poly A
(Promega) xEF1 a from pCI Luciferase
(Promega)
1736 pGL4 Complete Nil Photinus SV40 polyA and
(Promega) xEF1 a Luciferase enhancer
1689 pGL4 Complete Nil Photinus SV40 polyA
(Promega) xEF1 a Luciferase
1688 pGL4 Complete Rabbit Photinus SV40 polyA
(Promega) xEF1 a Pglobin Luciferase
1687 pGL4 Core Nil Photinus SV40 polyA
(Promega) xEF1 a Luciferase
1686 pGL4 Core Nil Photinus SV40 polyA
(Promega) xEF1 a Luciferase
1506 pGL4 Human Human Photinus SV40 polyA
(Promega) EF1 a EF1 alpha Luciferase
intron
1767 pKan Core Nil EGFP SV40 polyA and xEF1 a enhancer
1840 pKan Core Nil EGFP SV40 polyA and xEF1 a-TO enhancer
Examples
The examples are in three parts:
1 . Quantitative comparisons of promoter strengths in vitro.
2. Study of utility of selected promoter arrangements in adenovirus.
3. Description of promoter modifications which will also be of utility in adenovirus.
Example 1. Quantitative comparisons of promoter strengths in vitro
A series of reporter vectors were constructed, expressing Photinus luciferase under the control of a series of promoters. Xenopus EF1 a promoter variants were constructed by a combination of gene synthesis and conventional subcloning. As a comparator, we included the IE94 gene promoter from simian CMV, which is a strong promoter of similar function to the widely used human immediate early cytomegalovirus promoter. Sequence details are in Table 1 . These were transiently transfected into HeLa cells (ATCC CCL-2) or NIH-3T3 cells (ATCC CRL-1658), using an internal control HSV-TK expression plasmid (Promega, pGL4.74), using Superfect (Qiagen). 16-24 hours after transfection, reporter activity was assayed using a Dual Luciferase kit (Promega). Results were expressed as luciferase activities relative to that produced by pGL4.74.
Example 2. Study of utility of selected promoter arrangements in adenovirus
Construction and titration of adenoviruses
Bicistronic adenoviruses were constructed. A monocistronic adenoviral plasmid, AdC63. METRAP, was converted into a bicistronic plasmid expressing METRAP by addition of a cassette expressing the enhanced green fluorescent protein (EGFP, Clontech). The vector is illustrated schematically below (Figure 2). A shuttle plasmid (pKan) was constructed for this purpose. The pre-adenoviral AdC63. METRAP plasmid was linearised at the unique AsiSI restriction site between the Bovine Growth hormone polyadenylation signal and the adenoviral gene IX, and dephosphorylated. This was co-transformed into electrocompetent BJ5183 cells (Stratagene) along with a linearised fragment of pKan containing the EGFP expression cassette to be added, and homology arms identical to the adenoviral plasmid sequence adjacent to the unique site of linearisation. This process was essentially as described (Chartier, C, et al., J Virol, 1996. 70(7): p. 4805-10).
Viral Growth
Adenoviral plasmids were linearised, and 5mcg of DNA was transfected into 293 cells (ATCC CRL-1573) using Lipofectamine 2000 (Invitrogen), according to the manufacturer's recommendation. Cell growth media consisted of Dulbecco's Modified Eagle Medium (DMEM), supplemented by penicillin, streptomycin, 2mM L-glutamine and 10% fetal bovine serum (all form Life Technologies). Viruses were grown for five serial passages in 293 cells, and purified by Caesium chloride centrifugation after a varying number of passages. Quantitative PCR was performed genome copy number, and infectivity determined using anti- hexon immunostaining.
Viral stability determination
10Λ12χ vp of virus were purified as above, and completely digested with Pmel and Notl (New England Biolabs). The plasmids from which the adenoviruses were derived were cut in parallel. Fragments were resolved on 0.8% agarose gels.
Expression of METRAP antigen and GFP
293 cells were infected with AdC63 expressing METRAP, or METRAP together with EGFP under the control of the sCMV promoter, xEF1 promoter, or xEF1 promoter-SV40 enhancer at an MOI of 0.1 . 24 hours later, cells were fixed with 2% paraformaldehyde in 0.1 M Tris-HCI pH 8.0, and permeabilised with 0.1 M Tris-HCI pH 8.0, 0.05% Tween 20. Immunostaining was performed with a mouse anti-METRAP antiserum (a kind gift of Okairos AG, Italy) at 1 :200, and rabbit anti-mouse Alexa 647 (Invitrogen) was used as a second layer. Immunofluorescence images were obtained using an Eclipse microscope (Nikon), standard filters and a CCD camera (Hanamatsu).
Immunisation
Female Balb/c mice (Harlan UK Limited) were used in all experiments under the terms of the U.K. Home Office Animals (Scientific Procedures) Act Project Licence. Animals, 6-1 1 week of age, were immunised with 2.5χ10Λ7 ihu i.d., and γ-IFN ELISPOT assays performed on splenocytes 14 days later. A single cell suspension of splenocytes was generated mechanically, and erythrocytes lysed by resuspension of ACK buffer (0.15M NH4CI, 1 mM KHC03, 0.1 M Na2EDTA, pH 7.3), followed by resuspension in growth media (a-MEM, 10% FCS, 2mM L- glutamine, 0.1 % β-mercaptoethanol, 100U penicillin and 100ug streptomycin) (all from Life Technologies). Cells were plated on Multiscreen® filter plates (Milipore) pre-coated with mAbAN 18 (Mabtech) and stimulated for 18 to 20 h with ^g/ml Pb9 peptide (Kd-restricted CD8+ peptide SYIPSAEKI, a components of ME- TRAP) (Invitrogen, United Kingdom) or ^g/ml EGFP peptide. Cell concentrations were 2.5x106/ml (for Pb9) or 5x106/ml (for EGFP peptide). Plates were developed using mAb R4-6A2-biotin (Mabtech), Streptavidin-ALP (Mabtech), BIORAD AP-conjugate Substrate kit and enumerated using an ELISPOT Reader System ELR04 (AID GmbH, Strassberg, Germany). Assay methods have been previously described.
Example 3. Description of promoter modifications which will also be of utility in adenovirus
Two tetracycline operator sites were introduced downstream of the TATA box in xEF1 a by gene synthesis (Geneart AG) and subcloned promoters with and without the tet operators (sequences are in Table 1 ) into pKan upstream of EGFP. The resulting plasmids were transfected into 293 (ATCC) and 293-Trex (Invitrogen) cells using Superfect (Qiagen) as recommended by the manufacturer. 24 hour later, GFP fluorescence in the cells was determined by flow cytometry using a CyAn cytometer (Dako Cytomation). Results for Example 1.
Quantitative comparisons of promoter strengths in vitro
A series of vectors were constructed expressing Photinus luciferase under the control of a series of promoters. Some promoters contained natural or synthetic splice signals (Table 1 ). These included the human CMV immediate early cassette 94 promoter (hCMV), the human EF1 a promoter (hEF1 ), the simian CMV immediate early 94 promoter (sCMV), and the Xenopus EF1 a promoter. Some constructs also contained the SV40 enhancer. A schematic organisation of the various promoters tested is shown in Table 1. Numbers before each construct are plasmid identifiers. The activities shown are mean and standard deviation of three replicate transfections, in which 200ng of plasmid was co-transfected with 20ng of pGL4.74 (Promega, which expresses Renilla luciferase under the control of the TK promoter), and luciferase measurement were made with a Dual Luciferase assay (Promega).
Transient transfection of reporter constructs shows the Xenopus EF1 a promoter (xEF1 ) to be active in HeLa and 3T3 cells (Figure 1 ). Activity in vitro is significantly enhanced by the presence of the SV40 enhancer, and is comparable to that of the simian CMV promoter (Figure 1 ). Based on these data, adenoviruses were constructed using two of these promoter arrangements (Figure 2).
Viruses containing the xEF1 -alpha cassette are stable, but those containing sCMV are not stable
After serial passage, DNA was extracted from viruses containing the sCMV promoter, xEF1 promoter, or xEF1 promoter-SV40 enhancer. Restriction analysis showed the expected pattern from both xEF1 viruses, but the sCMV containing virus had changed during passage, compatible with inter-CMV promoter recombination (Figure 3). Results for Example 2.
Study of selected promoter arrangement in adenovirus
AdC63 adenoviruses expressing the METRAP antigen, and derivatives containing a second expression cassette (described above) were used to infect were recovered following growth in 293 cells. 293 cells were infected with AdC63 expressing METRAP, or METRAP together with EGFP under the control of the sCMV promoter, xEF1 promoter, or xEF1 promoter-SV40 enhancer at an MOI of 0.1. Under these conditions, virus is limiting, and infection of a single cell is likely to originate from one virion. Immunofluorescence microscopy was performed 24 hours after infection. Figure 2 shows representative cells from a large number examined; both METRAP and EGFP expression was demonstrable (Figure 4) in all cells, demonstrating function of the promoter, and viral stability.
The product of the xEF1 alpha cassette is immunogenic
2 weeks after immunisation with 1x10Λ7 infectious units of AdC63-METRAP- XEF1 -GFP, AdC63-METRAP-xEF1 -GFP-SV40, or AdC63-METRAP, spleen ELISPOT assays were performed using peptides present in ME-TRAP or EGFP. Responses are shown in Figure 5. Responses to METRAP are unaffected by the xEF1 -GFP-SV40 cassette, which generated immune responses to GFP.
Results for Example 3. Repressible versions of the promoter can be constructed
It is described that incorporation of tetracycline operator sites downstream of the human CMV promoter TATA box causes repression of transcription from the CMV promoter when tetracycline repressor proteins are present. An xEF1 a promoter containing this configuration (xEFLTO) was constructed. Activity analysis was performed by flow cytometry, monitoring EGFP expression (Figure 6). These show that the xEFLTO promoter has comparable activity to xEF1 when transfected into 293 cells, which lack the Tetracycline repressor protein. By contrast, substantial repression was seen when the same constructs were transfected into 293.tRex (Invitrogen) cells, in which the Tetracycline repressor is expressed.

Claims

Claims
1. A viral vector, comprising a first promoter sequence, wherein the first promoter sequence comprises:
(a) a nucleic acid sequence having at least 70% sequence identity to the nucleic acid sequence of SEQ ID NO: 1 , or
(b) at least two fragments of (a), wherein each fragment comprises at least 10 consecutive nucleotides of (a), or
(c) a fragment of (a), wherein said fragment comprises at least 20 consecutive nucleotides of (a).
2. The viral vector of claim 1 , further comprising an enhancer sequence; wherein the enhancer sequence increases transcription from the first promoter sequence.
3. The viral vector of claim 2, wherein the enhancer sequence comprises an SV40 enhancer.
4. The viral vector of claim 2, wherein the enhancer sequence comprises a Xenopus EF1 -alpha enhancer.
5. The viral vector of any of claims 1 -4, further comprising at least one gene; wherein the first promoter sequence is operably-linked to the at least one gene.
6. The viral vector of any of claims 1 -4, further comprising a second promoter sequence;
wherein the first promoter sequence and the second promoter sequence are different.
7. The viral vector of claim 6,
wherein the first promoter sequence is operably-linked to a first gene; and wherein the second promoter sequence is operably-linked to a second gene.
8. The viral vector of claim 6, wherein the second promoter sequence is selected from the group consisting of: human CMV promoter, simian CMV promoter, murine or other mammalian CMV promoters, promoters derived from other herpes viruses, promoters from other types of virus, and mammalian promoters.
9. The viral vector of any of claims 1 -8, wherein the viral vector is an adenovirus vector.
10. The viral vector of any of claims 1 -9, further comprising at least one operator site sequence.
1 1 . The viral vector of claim 10, wherein the at least one operator site sequence comprises a tetracycline operator.
12. A method of making a viral vector, comprising
providing a nucleic acid, wherein the nucleic acid comprises a nucleic acid sequence encoding a viral vector according to any of claims 1 -1 1 ;
transfecting a host cell with the nucleic acid;
culturing the host cell under conditions suitable for the expression of the nucleic acid; and
obtaining the viral vector from the host cell.
13. A host cell, comprising a viral vector according to any of claims 1 -1 1 .
14. An in vitro method of expressing in a target cell at least one protein, comprising
providing a viral vector according to any of claims 1-11; and
introducing the viral vector into a target cell.
15. A viral vector according to any of claims 1-11, for use in medicine.
16. A viral vector according to any of claims 1-11, for use in gene therapy.
17. A viral vector according to any of claims 1-11, for use in the treatment of cancer.
18. A viral vector according to any of claims 1-11, for use in the treatment of allergy.
19. A viral vector according to any of claims 1-11, for use in stimulating or inducing an immune response in a subject.
20. A viral vector according to any of claims 1-11, for use in the treatment or prevention of at least one infectious disease.
21. The viral vector for use according to claim 20, wherein the at least one infectious disease is selected from the group consisting of: malaria, influenza, and tuberculosis.
22. An immunological composition, comprising
a viral vector according to any of claims 1-11; and
a pharmaceutically acceptable carrier.
23. A pharmaceutical composition, comprising
a viral vector according to any of claims 1-11; and a pharmaceutically acceptable carrier.
24. A viral vector substantially as described herein with reference to the accompanying figures.
PCT/GB2011/050485 2010-03-12 2011-03-11 Promoter sequence for dna and viral vectors WO2011110864A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1004143.2 2010-03-12
GBGB1004143.2A GB201004143D0 (en) 2010-03-12 2010-03-12 Promoter sequence for DNA and viral vectors

Publications (1)

Publication Number Publication Date
WO2011110864A1 true WO2011110864A1 (en) 2011-09-15

Family

ID=42261483

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/050485 WO2011110864A1 (en) 2010-03-12 2011-03-11 Promoter sequence for dna and viral vectors

Country Status (2)

Country Link
GB (1) GB201004143D0 (en)
WO (1) WO2011110864A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013061010A1 (en) * 2011-10-28 2013-05-02 Lfb Biotechnologies Transcription unit and use thereof in (yb2/0) expression vectors
WO2017132376A1 (en) * 2016-01-27 2017-08-03 Just Biotherapeutics, Inc. Hybrid promoter and uses thereof
CN109468323A (en) * 2018-11-21 2019-03-15 新乡医学院 Artificial synthesized introne, mammalian cell recombinant expression carrier, mammalian host cell, expression and its application
US11098310B2 (en) 2016-01-27 2021-08-24 Just-Evotec Biologics, Inc. Expression from transposon-based vectors and uses
US11261462B2 (en) 2016-01-27 2022-03-01 Just-Evotec Biologics, Inc. Inducible expression from transposon-based vectors and uses

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1004143A (en) 1960-09-26 1965-09-08 William Edward O Shei Improvements in or relating to vacuum die-casting
WO2002088368A1 (en) * 2001-05-02 2002-11-07 Institute Of Molecular Agrobiology Spatial and temporal control of gene expression in the zebrafish using baculovirus
US20060063727A1 (en) * 2002-05-17 2006-03-23 Chawnshang Chang Tr4/tr2 response elements
US20090099067A1 (en) * 2006-03-17 2009-04-16 Avigenics, Inc. Glycosylated G-CSF

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1004143A (en) 1960-09-26 1965-09-08 William Edward O Shei Improvements in or relating to vacuum die-casting
WO2002088368A1 (en) * 2001-05-02 2002-11-07 Institute Of Molecular Agrobiology Spatial and temporal control of gene expression in the zebrafish using baculovirus
US20060063727A1 (en) * 2002-05-17 2006-03-23 Chawnshang Chang Tr4/tr2 response elements
US20090099067A1 (en) * 2006-03-17 2009-04-16 Avigenics, Inc. Glycosylated G-CSF

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
BELOUSOVA, N. ET AL., MOL PHARMACOL, vol. 70, no. 5, 2006, pages 1488 - 93
CAYER, M.P. ET AL., J IMMUNOL METHODS, vol. 322, no. 1-2, 2007, pages 118 - 27
CHARTIER, C. ET AL., J VIROL, vol. 70, no. 7, 1996, pages 4805 - 10
GONZALEZ-NICOLINI, V.; M. FUSSENEGGER, METHODS MOL BIOL, vol. 434, 2008, pages 221 - 37
JOHNSON A D ET AL: "pXeX, a vector for efficient expression of cloned sequences in Xenopus embryos", GENE, ELSEVIER, AMSTERDAM, NL, vol. 147, no. 2, 30 September 1994 (1994-09-30), pages 223 - 226, XP023324916, ISSN: 0378-1119, [retrieved on 19940930], DOI: DOI:10.1016/0378-1119(94)90070-1 *
LIMBACH K J ET AL: "Viral vectors in malaria vaccine development", PARASITE IMMUNOLOGY, BLACKWELL SCIENTIFIC PUBLICATIONS, OXFORD, GB, vol. 31, no. 9, 1 September 2009 (2009-09-01), pages 501 - 519, XP002601705, ISSN: 0141-9838, [retrieved on 20090626], DOI: DOI:10.1111/J.1365-3024.2009.01141.X *
SUDOWE S ET AL: "Efficacy of recombinant adenovirus as vector for allergen gene therapy in a mouse model of type I allergy", GENE THERAPY, vol. 9, no. 2, January 2002 (2002-01-01), pages 147 - 156, XP002634211, ISSN: 0969-7128 *
TENBUSCH, M. ET AL., BMC IMMUNOL, vol. 9, 2008, pages 13
VOGELS, R. ET AL., J GEN VIROL, vol. 88, 2007, pages 2915 - 24
YOON KEEJUNG ET AL: "Fibroblast growth factor receptor signaling promotes radial glial identity and interacts with Notch1 signaling in telencephalic progenitors", JOURNAL OF NEUROSCIENCE, vol. 24, no. 43, 27 October 2004 (2004-10-27), pages 9497 - 9506, XP002634210, ISSN: 0270-6474 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013061010A1 (en) * 2011-10-28 2013-05-02 Lfb Biotechnologies Transcription unit and use thereof in (yb2/0) expression vectors
FR2981946A1 (en) * 2011-10-28 2013-05-03 Lfb Biotechnologies TRANSCRIPTION UNITS AND THEIR USE IN EXPRESSION VECTORS (YB2 / 0)
US9551009B2 (en) 2011-10-28 2017-01-24 Laboratoire Francais Du Fractionnement Et Des Biotechnologies Transcription unit and use thereof in expression vectors
WO2017132376A1 (en) * 2016-01-27 2017-08-03 Just Biotherapeutics, Inc. Hybrid promoter and uses thereof
JP2019503179A (en) * 2016-01-27 2019-02-07 ジャスト バイオセラピューティックス,インコーポレイテッド Hybrid promoter and use thereof
US11028410B2 (en) 2016-01-27 2021-06-08 Just-Evotec Biologics, Inc. Hybrid promoter and uses thereof
US11098310B2 (en) 2016-01-27 2021-08-24 Just-Evotec Biologics, Inc. Expression from transposon-based vectors and uses
US11261462B2 (en) 2016-01-27 2022-03-01 Just-Evotec Biologics, Inc. Inducible expression from transposon-based vectors and uses
US11685933B2 (en) 2016-01-27 2023-06-27 Just-Evotec Biologics, Inc. Inducible expression from transposon-based vectors and uses
US11692193B2 (en) 2016-01-27 2023-07-04 Just-Evotec Biologies, Inc. Expression from transposon-based vectors and uses
CN109468323A (en) * 2018-11-21 2019-03-15 新乡医学院 Artificial synthesized introne, mammalian cell recombinant expression carrier, mammalian host cell, expression and its application
CN109468323B (en) * 2018-11-21 2022-03-11 新乡医学院 Artificially synthesized intron, mammalian cell recombinant expression vector, mammalian host cell, expression method and application thereof

Also Published As

Publication number Publication date
GB201004143D0 (en) 2010-04-28

Similar Documents

Publication Publication Date Title
KR101377582B1 (en) Dna modular cloning vector plasmids and methods for their use
KR101724162B1 (en) Gene therapy vectors and cytosine deaminases
US5952171A (en) Method for identifying genes encoding secreted or membrane-associated proteins
US20040185556A1 (en) DNA cloning vector plasmids and methods for their use
CN110079551B (en) Circular RNA expression vector and construction method and application thereof
WO2011110864A1 (en) Promoter sequence for dna and viral vectors
CN113831394B (en) Recombinant virus combination of African swine fever virus ASFV gene and vaccine prepared from recombinant virus combination
CA2318575A1 (en) Feline immunodeficiency virus gene therapy vectors
US20030077251A1 (en) Replicons derived from positive strand RNA virus genomes useful for the production of heterologous proteins
CN106755100A (en) The genomes of controllability HIV 1 target editing system and its targeting carrier system
US6573100B1 (en) DNA vectors without a selection marker gene
CN110257403B (en) Infectious laryngotracheitis virus gB gene expression, recombinant fowlpox virus thereof, construction method and application
CN111110707A (en) Application of recombinant oncolytic virus in preparation of medicine for treating digestive tract tumor
CN104328140B (en) A kind of high preferendum bladder cancer target tumor-killing adenovirus
CN113880957A (en) Streptolysin O fusion protein
CN115403666A (en) Anti-influenza virus polypeptide and application thereof
CN114835822B (en) Polymer vaccine of hog cholera virus and its preparing process
CN114457118B (en) Fluorescent reporter gene element, gene editing and monitoring system and application thereof
CN111440772A (en) Anti-human papilloma virus drug screening model and construction method and application thereof
CN111454962B (en) Fixed-point modification based on bovine safe site and application thereof
CN112442512A (en) Gene editing system for Japanese medaka embryos and cells based on tRNA-gRNA-cRNA
CN112410344B (en) CpG-ODN with specific immunostimulation effect on PRRSV and application thereof
CN109609554B (en) Cord mesenchymal stem cell with mir338 gene silencing function and preparation method and application thereof
WO2013164792A1 (en) Novel transgenic animal model of amyotrophic lateral sclerosis
CN115521364A (en) SARS-CoV-2 delta mutant strain S protein variant and its use

Legal Events

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

Ref document number: 11709174

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11709174

Country of ref document: EP

Kind code of ref document: A1