EP0833896A1 - Rekombinante adenovirus,ihre verwendung zur herstellung von aav,zur komplementation falige zellinie und diese enthaltende pharmazeutische zusammensetzungen - Google Patents

Rekombinante adenovirus,ihre verwendung zur herstellung von aav,zur komplementation falige zellinie und diese enthaltende pharmazeutische zusammensetzungen

Info

Publication number
EP0833896A1
EP0833896A1 EP96922971A EP96922971A EP0833896A1 EP 0833896 A1 EP0833896 A1 EP 0833896A1 EP 96922971 A EP96922971 A EP 96922971A EP 96922971 A EP96922971 A EP 96922971A EP 0833896 A1 EP0833896 A1 EP 0833896A1
Authority
EP
European Patent Office
Prior art keywords
promoter
adenovirus according
tetracycline
recombinant adenovirus
region
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP96922971A
Other languages
English (en)
French (fr)
Inventor
Martine Latta
Cécile Orsini
Michel Perricaudet
Edouard Prost
Emmanuelle Vigne
Patrice Yeh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aventis Pharma SA
Original Assignee
Rhone Poulenc Rorer SA
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 Rhone Poulenc Rorer SA filed Critical Rhone Poulenc Rorer SA
Publication of EP0833896A1 publication Critical patent/EP0833896A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • 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
    • C12N2710/10344Chimeric viral vector comprising heterologous viral elements for production of another viral vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16622New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use 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
    • C12N2830/002Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor
    • C12N2830/003Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor tet inducible
    • 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

Definitions

  • the present invention relates to new viral vectors, their preparation and their uses. It also relates to pharmaceutical compositions containing said viral vectors.
  • Gene therapy consists of correcting a deficiency or an anomaly (mutation, aberrant expression, etc.) by introducing genetic information into the affected cell or organ.
  • This genetic information can be introduced either in vitro into a cell extracted from the organ, the modified cell then being reintroduced into the organism, or directly in vivo into the appropriate tissue.
  • different techniques exist, among which various transfection techniques involving complexes of DNA and DEAE-dextran (Pagano et al., J. Virol.
  • viruses as vectors for gene transfer has emerged as a promising alternative to these physical transfection techniques.
  • different viruses have been tested for their ability to infect certain cell populations.
  • retroviruses RSV, HMS, MMS, etc.
  • the HSV virus adeno-associated viruses
  • adenoviruses adenoviruses
  • viruses with linear double strand DNA with a size of approximately 36 kb.
  • Their genome includes in particular an inverted sequence (ITR) at each end, an encapsidation sequence, early genes and late genes (see FIG. 1).
  • the main early genes are contained in the E1, E2, E3 and E4 regions.
  • the genes contained in the El region are necessary for viral replication.
  • the E4 and L5 regions for example are involved in the viral spread and the main late genes are contained in regions LI to L5.
  • the genome of the Ad5 adenovirus has been fully sequenced and is accessible on the database (see in particular Genebank M73260).
  • adenoviruses of different serotypes have also been sequenced.
  • These viral vectors advantageously have a fairly broad host spectrum, are capable of infecting quiescent cells, do not integrate into the genome of the infected cell, and have not been associated to date with significant pathologies in humans. 'man. Given their properties, they have already been used for gene transfer in vivo.
  • different vectors derived from adenoviruses have been prepared, incorporating different genes ( ⁇ -gal, OTC, ⁇ -lAT, cytokines, etc.).
  • the viral vector constructions currently proposed are modified so as to render said vectors incapable of replicating autonomously in the target cell. They are said to be defective.
  • the genome of defective viruses is therefore devoid of at least the sequences necessary for the replication of said virus in the infected cell. These regions can be either eliminated (in whole or in part), or made non-functional, or substituted by other sequences and in particular by a sequence coding for a molecule of therapeutic interest.
  • the defective virus nevertheless retains the sequences of its genome which are necessary for the packaging of the viral particles.
  • the constructions described in the prior art are generally adenoviruses deleted from the El (El a and / or Elb) and possibly E3 regions into which the heterologous DNA sequences are inserted (Levrero et al ., Gene 101 (1991) 195; Gosh-Choudhury et al., Gene 50 (1986) 161).
  • Other constructions include deletion at the level of the E1 region and a non-essential part of the E4 region (WO 94/12649).
  • adenovirus serotype 5 Ad5
  • This line is capable of trans-complementing recombinant adenoviruses defective for the E1 region, that is to say devoid of all or part of the E1 region, necessary for replication.
  • replicative viral particles can have very harmful consequences such as for example inducing a viral propagation and causing an uncontrolled dissemination with risks of inflammatory reaction, recombination, etc ...
  • the present invention relates to a recombinant adenovirus in which the expression of at least one gene of viral origin, homologous or heterologous, is controlled by an inducible promoter.
  • inducible promoter means any promoter whose activity is initiated by the presence of an external chemical and / or biological agent, an agent which, in the context of the present invention also has reduced toxicity or even zero.
  • exterior is meant that the chemical and / or biological agent does not naturally exist in cells treated with the claimed adenovirus.
  • inducible promoters capable of being used according to the present invention, mention may in particular be made of conventional promoters such as those responding to heavy metals (CRC Boca Raton, FL (1991), 167-220; Brinster et al.
  • a promoter inducible by tetracycline comprises a minimal promoter operably linked to one or more tetracycline operator (s). It is the binding, to the tetracycline operator sequences, of a protein called "transcription activator", a binding which is established only in the presence of tetracycline or one of its analogs, which allows the activation of the minimal promoter and therefore the transcription of the associated viral gene or genes.
  • transcription activator a binding which is established only in the presence of tetracycline or one of its analogs, which allows the activation of the minimal promoter and therefore the transcription of the associated viral gene or genes.
  • transcription activator it is therefore characterized by its ability to bind, in the presence of tetracycline, to the operator sequences of the promoter inducible by tetracycline and its capacity to activate the minimal promoter. More preferably, it is a protein consisting of two polypeptides, a first polypeptide which binds to the operator tet sequences, in the presence of tetracycline or an analog thereof, and a second polypeptide whose function is more specifically to activate said transcription.
  • the first polypeptide of the protein known as transcription activator is a repressor of tetracycline mutated so as to manifest a behavior opposite to that of a wild repressor, that is to say that it binds to the tet operator sequences only in the presence and not in the absence of tetracycline.
  • the second polypeptide it is preferably the activation domain of protein 16 of the Herpes Simplex Virus.
  • the inducible promoter used is, for example, inducible to glucose or galactose, it is possible to envisage using a transcription activator constructed on this model, that is to say for example Glu-VP16 or Gal4-VP16.
  • the inducible promoter used is a promoter inducible to tetracycline or one of its analogs as described above.
  • a promoter inducible by tetracycline comprises a minimal promoter operably linked to a so-called regulatory sequence comprising at least one operator of the tetracycline "tet operator" or one of its analogs.
  • tetracycline analog is intended to cover any compound having structural homologies with tetracycline and being capable of binding to its receptor linked to the transactivation domain of the protein called trancription activator presented above, with a Ka of at least. at least about 10 6 M " 1.
  • analogs capable of being used according to the present invention mention may in particular be made of doxycycline, chlorotetracycline and ranhydrotetracycline.
  • minimal promoter any promoter sequence which alone is not capable of efficiently ensuring the transcription of the DNA sequence which is associated with it.
  • the activity of such a promoter turns out to be totally dependent on the binding of the transcription activator protein to the so-called regulatory sequence, in the presence of tetracycline.
  • this minimal promoter mainly has the function of directing transcription. In this perspective, it is preferably located upstream of the viral sequence so as to form with it a continuous nucleotide sequence.
  • This minimal promoter can be derived from the immediately early promoter of human Cytomegalovirus and more preferably is between nucleotides +75 to -53 or +75 to -31.
  • a minimal promoter deriving from a conventional promoter such as for example that activating the transcription of the gene coding for thymidine kinase.
  • a conventional promoter can also be minimized through one or more genetic mutations which render it incapable of ensuring efficient transcription of the gene associated with it alone. It can also be used in the context of the present invention, a minimal promoter deriving directly from the promoter naturally responsible for the expression of the viral gene considered.
  • a promoter called "TATA-less” as described by E. MARTINEZ et al. (EMBO Journal, (1994), 13, N ° 13, 3115-3126) so as to obtain the most basic background noise possible in the non-induced situation.
  • this irunirnal promoter is placed upstream of the nucleotide sequence the expression of which it controls, whether or not in substitution for its natural promoter.
  • the promoter of the nucleic acid sequence can in fact remain present but in an inactivated form or made non-functional by various techniques known to those skilled in the art and in particular by deletion, deletion and / or addition of one or more bases.
  • the ierinimal promoter is derived from the minimal promoter of the thymidine kinase of the Herpes Simplex Virus (Me Knight et al. (1984) Cell 37: 253-262). He is then designated by Tk. More preferably, it is represented in whole or in part by one of the sequences represented in SEQ ID No. 1 or No. 2 or one of their derivatives.
  • the term derivative designates any sequence obtained by modification of a genetic and / or chemical nature of given sequences and retaining the desired activity.
  • modification of a genetic and / or chemical nature is meant any mutation, substitution, deletion, addition and / or modification of one or more nucleic acid.
  • the so-called regulatory sequence comprises at least one operator of tetracycline or one of its analogs.
  • the operator (s) are recognized by the transcription activator in the presence of tetracycline and therefore allow the activation of the minimal promoter.
  • the tet operator sequences which can be implemented can in particular be chosen from those described by Hillen & Wissemann (Protein-Nucleic Acid Interaction, Saeger & Heinemann, eds., Macmillan, London, (1989), 10, 143-162), Waters and al. (Nucleic Acids Res. (1983), 11, 525-539), Stuber et al. (P.N.A.S. USA, (1981), 78, 167-171), Unger et al. (Nucleic Acids Res. (1984), 12, 7693-7703) and Tovar et al. (Mol. Gen. Genêt. (1988), 215, 76-80).
  • the regulation sequence can comprise a single operator tet sequence or on the contrary several operator tet sequences up to number of 10 depending on whether or not it is desired to increase the regulation of transcription. According to a particular embodiment of the invention, the regulation sequence implements 2 operator tet sequences. It will then be called Op2.
  • the regulatory sequence is represented in whole or in part by one of the sequences represented in SEQ ID No. 3 or No. 4 or one of their derivatives.
  • this regulatory sequence is operatively linked upstream, that is to say at the 5 ′ end of the minimal promoter so as to allow transcription of the gene of viral origin, in the presence of the complex formed by the activator of transcription and its tetracycline ligand.
  • the regulatory sequence linked or not directly to the minimal promoter, the minimal promoter and the gene of viral origin.
  • this regulatory sequence within the minimal promoter, downstream of the viral nucleotide sequence to be transcribed, that is to say at its 3 ′ end. The order of succession is then in the 5 ′ to 3 ′ direction, minimal promoter, viral gene and regulatory sequence.
  • the promoter inducible to tetracycline combines with the minimal promoter of thymidine kinase called Tk, a regulatory sequence represented by Op2. In this particular case, it is identified below under the name Op2 / Tk. More preferably, the inducible promoter used according to the invention is represented in whole or in part by SEQ ID No. 5 or one of its derivatives.
  • This promoter inducible to tetracycline Op2 / Tk and more particularly that represented in whole or in part by SEQ ID No. 5 or one of its derivatives, also constitute one of the objects of the present invention.
  • the expression of the viral gene (s) operatively linked, in the claimed adenovirus, to an inducible promoter is totally subject to the binding of the complex formed by the transcription activator and the tetracycline, on the regulatory sequence of said promoter.
  • One of the objects of the present invention relates more particularly to an adenovirus comprising at least one homologous gene, that is to say adenoviral, the expression of which is controlled by an inducible promoter and more preferably by a promoter inducible to tetracycline.
  • the subject of the present invention is a recombinant adenovirus of which at least one genomic region essential for replication and / or viral propagation is placed in whole or in part under the control of a promoter inducible to tetracycline.
  • the region essential for replication and / or viral propagation according to the present invention is advantageously chosen from all or part of the E4 region, E2, the IVa2 region and / or the L5 region, etc.
  • the recombinant adenoviruses of the present invention comprise, as sequences necessary for replication and / or propagation, all or a functional part of the E2 or E4 regions. More particularly, with regard to the E4 region, the important genes are the ORF3, ORF6 and ORF6 / 7 genes.
  • the E2 region is involved in the regulation of viral DNA.
  • This E2 region consists of two E2A and E2B transcription subunits.
  • the E4 region is involved in the regulation of late gene expression, in the stability of late nuclear RNA, in the quenching of expression of host cell proteins and in the efficiency of DNA replication viral. Mutants lacking E4 are unable to spread. E4 thus constitutes a region essential for viral spread.
  • This E4 region consists of 7 open reading phases, designated ORF1, ORF2, ORF3, ORF4, ORF3 / 4, ORF6 and ORF6 / 7 ( Figure 2).
  • ORF3 and ORF6 are the two genes essential for viral spread. Each of these genes is capable of inducing viral propagation, however ORF6 plays a more important role there than ORF3 (Huang and Hearing (1988), J. Virol. 63, 2605).
  • the entire region considered is placed under the control of a promoter inducible to tetracycline.
  • the E2 region may be a fragment corresponding to the 72K cDNA, to the 140K polymerase cDNA or to the 87K pre-terminal protein cDNA.
  • the E4 region it may in particular be the Taql-Bgl2 fragment corresponding to nucleotides 35576 -32490.
  • this part comprises at least one functional ORF3 or ORF6 gene.
  • the functional part of E4 consists essentially of ORF6.
  • the Bgl2 fragment, lying between positions 34115 to 32490 and containing the sequences of ORF6 and ORF7 of Ad5 can be positioned downstream of an inducible promoter as defined according to the invention.
  • the essential region consists of the region coding for the protein IVa2 and for example its cDNA.
  • the region coding for the protein IVa2 is included in a BglII-NruI fragment corresponding to nucleotides 3328 to 6316 on the sequence of the wild-type Ad5 adenovirus, a Dral-NlalII fragment corresponding to nucleotides 4029 to 5719 or a Xhal dral fragment corresponding to nucleotides 4029 to 5788.
  • the promoters of the regions essential for viral propagation are replaced within the viral genome by an inducible promoter and more preferably by a promoter inducible to tetracycline.
  • the recombinant adenoviruses of the invention carry a deletion of all or part of the El gene and have the E4 region, in whole or in part, under the control of a promoter inducible to tetracycline, preferably type Op2 / Tk.
  • the recombinant adenoviruses of the invention carry a deletion of all or part of the E1 gene and have the E2 region in whole or in part under the control of a promoter inducible to tetracycline, preferably of the Op2 type / rk.
  • the recombinant adenoviruses of the invention carry a deletion of all or part of the El and E2 genes and have the E4 region wholly or partly under the control of a promoter inducible to tetracycline, preferably of type Op2 / Tk.
  • the recombinant adenoviruses of the invention carry a deletion of all or part of the El and E4 genes and have the E2 region in whole or in part under the control of a promoter inducible to tetracycline, preferably of the Op2 type / Tk.
  • the recombinant adenoviruses of the invention also comprise a heterologous nucleic acid sequence comprising one or more therapeutic genes whose transfer and / or expression in a cell, an organ or an organism is sought.
  • the therapeutic genes which can thus be transferred are any gene whose transcription and possibly translation into the target cell generate products having a therapeutic effect.
  • the protein product thus coded can be a protein, a peptide, etc.
  • This protein product can be homologous with respect to the target cell (that is to say a product which is normally expressed in the target cell when the latter has no pathology).
  • the expression of a protein makes it possible, for example, to compensate for an insufficient expression in the cell or the expression of a protein inactive or weakly active due to a modification, or to overexpress said protein.
  • the therapeutic gene can also code for a mutant of a cellular protein, having increased stability, modified activity, etc.
  • the protein product can also be heterologous towards the target cell.
  • an expressed protein can for example supplement or provide a deficient activity in the cell allowing it to fight against a pathology.
  • trophic factors BDNF, CNTF, NGF, IGF, GMF, aFGF, bFGF, NT3, NT5, etc; apolipoproteins: ApoAI, ApoAIV, ApoE, etc (FR 93 05125), dystrophin or a mini dystrophin (FR 9111947), tumor suppressor genes: p53, Rb, RaplA, DCC, k-rev, etc (FR 93 04745 ), genes coding for factors involved in coagulation: Factors VII, VIII, IX, etc., suicide genes: Thymidine kinase, cytosine deaminase, etc; or all or part of a natural or artificial immunoglobin, etc.
  • the therapeutic gene can also be an antisense gene or sequence, the expression of which in the target cell makes it possible to control the expression of genes or the transcription of cellular mRNAs such as ribozymes.
  • Such sequences can, for example, be transcribed, in the target cell, into RNAs complementary to cellular mRNAs and thus block their translation into protein, according to the technique described in patent EP 140 308.
  • the therapeutic gene can also be a gene coding for an antigenic peptide capable of generating an immune response in humans.
  • the invention therefore allows the production of vaccines making it possible to immunize humans, in particular against microorganisms or viruses.
  • These may in particular be antigenic peptides specific for the epstein barr virus, the HIV virus, the hepatitis B virus (EP 185 573), the pseudo-rabies virus, or even specific for tumors (EP 259 212).
  • the heterologous nucleic acid sequence also comprises a promoter region for functional transcription in the infected cell, as well as a region located 3 ′ of the gene of interest, and which specifies a transcriptional end signal and a site for polyadenylation.
  • the promoter region it may be a promoter region which is naturally resoonsable for the expression of the gene considered when it is capable of functioning in the infected cell. They can also be regions of different origin (responsible for the expression of other proteins, or even synthetic). In particular, they may be promoter sequences of eukaryotic or viral genes. For example, they may be promoter sequences originating from the genome of the cell which it is desired to infect. Likewise, they may be promoter sequences originating from the genome of a virus, including the adenovirus used. In this regard, mention may be made, for example, of the promoters of the E1A, MLP, CMV, RSV, etc. genes.
  • these promoter regions can be modified by adding activation or regulatory sequences, or allowing tissue-specific or majority expression.
  • the heterologous nucleic acid does not contain promoter sequences, it can be inserted into the genome of the defective virus downstream of such a sequence.
  • heterologous nucleic acid sequence may also comprise, in particular upstream of the therapeutic gene, a signal sequence directing the therapeutic product synthesized in the secretory pathways of the target cell.
  • This signal sequence may be the natural signal sequence of the therapeutic product, but it may also be any other functional signal sequence, or an artificial signal sequence.
  • This nucleic acid sequence is preferably present at the E1, E3 or E4 regions, in addition to or in replacement of deleted sequences.
  • the second main object of the present invention is an adenovirus comprising at least one gene of heterologous viral origin, the expression of which is controlled by an inducible promoter and more preferably a promoter inducible to tetracycline.
  • the gene of heterologous viral origin is or is derived from a gene of the genome of an AAV or one of its functional counterparts.
  • AAVs are relatively small DNA viruses that integrate into the genome of the cells they infect in a stable and site-specific manner. They are also capable of infecting a broad spectrum of cells, without inducing an effect on cell growth, morphology or differentiation. Furthermore, they do not seem to be involved in pathologies in humans.
  • the AAV genome has been cloned, sequenced and characterized. It comprises 4680 bases, and contains at each end an inverted repeat region (ITR) of approximately 145 bases, serving as an origin of replication for the virus.
  • ITR inverted repeat region
  • the rest of the genome is divided into 2 essential regions carrying the packaging functions: the left part of the genome, which contains the rep gene involved in viral replication and the expression of viral genes; the right part of the genome, which contains the eap gene coding for the capsid proteins of the virus.
  • Three promoters were located there and named according to their approximate position in map units p5, pl9 and p40. At least four proteins are synthesized from the rep region and have been named after their apparent molecular mass, Re ⁇ 78, Rep68, Rep52 and Rep40.
  • the 2 mRNAs transcribed from the p5 promoter are used for the synthesis of Rep78 and Rep68.
  • Rep52 and Rep40 they are synthesized from the messengers originating from the promoter pl9.
  • cap gene codes for the proteins of the capsid of the virus (VP1, VP2 and VP3).
  • VP3 is the majority capsid protein and its amino acid sequence is contained in those of two larger but less abundant proteins VP1 and VP2 (draw a diagram).
  • rep genes . and cjrj have been characterized and their respective sequences described in the literature (Srivastava et al., J. Virol. 45 (1983) 555).
  • vectors derived from AAVs for gene transfer in vitro and in vivo has been described in the literature (see in particular WO 91/18088; WO 93/09239; US 4,797,368, US 5,139,941, EP 488,528).
  • the constructions used in gene therapy contain a deletion of the rep and / or cjp genes which are replaced by a gene of interest.
  • AAVs need the presence of a helper virus capable of transcomplementing the functions necessary for their replication. It may in particular be an adenovirus, a herpes virus or a vaccinia virus. (In the absence of such a helper virus, the AAVs remain in latent form in the genome of the infected cells, but cannot replicate and thus produce viral particles.)
  • a helper virus for example an adenovirus
  • the recombinant AAVs are therefore produced by co-transfection , in a cell line infected with a human helper virus (for example an adenovirus), of a plasmid containing the gene of interest bordered by two inverted repeat regions (ITR) of AAV and of a plasmid carrying the genes of encapsidation (rep and cap genes) of AAV.
  • Co-infection with adenovirus starts a cascade of events that lead to the production of high AAV titers and significantly decrease the production of adenovirus.
  • This cascade starts with the synthesis of the Ela gene product which induces transcription from the p5 and p19 promoters and leads to the synthesis of a small amount of Rep proteins.
  • One or more Rep proteins synthesized from p5 then induce the synthesis in a more abundant quantity of mRNA from the 3 promoters at a much greater level and in a coordinated manner.
  • the AAV genome is either lost or integrated into the host chromosome.
  • Other genes than El A of the adenovirus are also necessary for efficient expression of the AAV genes.
  • the Applicant has demonstrated that it was possible to effectively place at least the expression of one of the AAV viral genes under the control of an inducible promoter in an adenovirus and more preferably to control the expression of AAV packaging functions, in particular the expression of the rep and / or cap genes, or of any functional homologous gene.
  • a functional counterpart corresponds to any gene obtained by modification (mutation, deletion, addition, etc.) of the rep or cap genes and having an activity of the same nature.
  • Such functional homologous genes can also be genes obtained by hybridization from nucleic acid libraries by means of probes corresponding to the rgp or cap genes.
  • a mutated re_p gene capable of being controlled according to the invention one can more particularly cite its mutant in1177 described in the publication Y. Yang et al., ((1992) Journal of virology, 6058-6069) and derived from '' insertion of serines between codons 286 and 287.
  • the inducible promoter used is a promoter inducible to tetracycline as defined above.
  • Such an adenovirus is advantageous for several reasons: it considerably simplifies, in terms of handling, the process for preparing stocks of AAV. Indeed, in this particular case, it essentially implements that said adenovirus comprising the rep and cap genes under the control of the inducible promoter, a recombinant AAV and an adequate cell line. Finally, the titers expected in AAV from such an adenovirus prove to be higher than those obtained according to a conventional process.
  • the inducible promoter can in particular be introduced in substitution for one of the promoters which normally lead to the expression of the gene (s) considered and in particular in substitution for the promoter p5, pl9 or p40.
  • the p5 promoter seems to be the most involved in the start of the cascade of events leading to the production of the virus, its substitution is more preferably carried out by a promoter inducible by tetracycline, preferably of the Op2 / Tk type.
  • tetracycline preferably of the Op2 / Tk type.
  • the AAV packaging functions under the control of the inducible promoter can be introduced into different regions of the genome of the claimed adenovirus.
  • the packaging functions are inserted in a region which does not disturb the ability of the virus to transcomplement the AAVs. It is also possible to insert the packaging functions into a functional region of the genome of said adenovirus, which region is then supplied in trans, either by a plasmid or by the cell line used. It is possible by example of inserting the rgp gene, the cjp gene or the rsp and cjp genes at the level of the El or E3 regions to replace or supplement the deleted sequences.
  • a sequence known as a negative regulatory sequence Such a sequence inserted in particular between the left ITR and the psi sequence of the claimed adenovirus on the one hand, and the sequence coding for the tetracycline-inducible promoter makes it possible to stem any parasitic transcriptional activation of rep and cap, induced if necessary by the enhancer located in the left ITR of the adenovirus and the psi sequence. Mention may in particular be made, as negative sequences which can be used according to the invention, of those identified in the vimentin promoter (Salvetti et al., (1993), Mol. Cell.
  • Biol., 1676-1685 in the interferon promoter (Whitemore et al. (1990), PNAS, 87, 7799-7803), in the light chain gene 2 of cardiac myosin (Ruoquian-Shen et al. (1991), Mol. Cell. Biol., 1676-1685 ) and in the mouse albumin promoter (Herbst et al. (1990), Mol. Cell. Biol., 3896-3905).
  • the invention relates to a recombinant adenovirus carrying an Op2 / Tk-rep-cap expression cassette.
  • the present invention also relates to the use of these adenoviruses integrating a viral sequence of AAV origin under the control of a promoter inducible to tetracycline to prepare AAVs.
  • the adenoviruses which are the subject of the invention comprise the ITR sequences and a sequence allowing the packaging.
  • these adenoviruses also have a non-functional E1 region.
  • the inverted repeat sequences constitute the origin of replication of adenoviruses. They are located at the 3 ′ and 5 ′ ends of the viral genome (cf. FIG. 1), from which they can be easily isolated according to the conventional techniques of molecular biology known to those skilled in the art.
  • the nucleotide sequence of the sequences are located at the 3 ′ and 5 ′ ends of the viral genome (cf. FIG. 1), from which they can be easily isolated according to the conventional techniques of molecular biology known to those skilled in the art.
  • ITR of human adenoviruses (in particular serotypes Ad2 and Ad5) is described in the literature, as well as canine adenoviruses (in particular CAV1 and CAV2).
  • Ad5 adenovirus for example, the left ITR sequence corresponds to the region comprising nucleotides 1 to 103 of the genome.
  • the packaging sequence (also called Psi sequence) is necessary for the packaging of viral DNA. This region must therefore be present to allow the preparation of defective recombinant adenoviruses according to the invention.
  • the packaging sequence is located in the genome of the adenoviruses, between the left ITR (5 ′) and the El gene (see FIG. 1). It can be isolated or artificially synthesized by conventional molecular biology techniques.
  • the nucleotide sequence of the packaging sequence of human adenoviruses (in particular the Ad2 and Ad5 serotypes) is described in the literature, as well as canine adenoviruses (in particular CAV1 and CAV2).
  • the packaging sequence corresponds to the region comprising nucleotides 194 to 358 of the genome.
  • the E1 region is inactivated by deletion of a PvuII-BglII fragment ranging from nucleotide 454 to nucleotide 3328, on the sequence of the adenovirus Ad5. This sequence is accessible in the literature and also on the database (see in particular Genebank n ° M73260).
  • the E1 region is inactivated by deletion of a HinfII-Sau3A fragment going from nucleotide 382 to nucleotide 3446.
  • adenoviruses of the invention can be prepared from adenoviruses of various origins. There are indeed various adenovirus serotypes, whose structure and properties vary somewhat, but which have a comparable genetic organization. Thus, the lessons described in the present application can be easily reproduced by those skilled in the art for any type of adenovirus.
  • the adenoviruses of the invention can be of human, animal, or mixed (human and animal) origin. Concerning adenoviruses of human origin, it is preferred to use those classified in group C. More preferably, among the various serotypes of human adenovirus, it is preferred to use, within the framework of the present invention, adenoviruses of type 2 or 5 (Ad 2 or Ad 5).
  • the adenoviruses of the invention can also be of animal origin, or contain sequences derived from adenoviruses of animal origin.
  • the Applicant has indeed shown that adenoviruses of animal origin are capable of infecting human cells with great efficiency, and that they are unable to propagate in the human cells in which they have been tested (cf. application WO 94 / 26914).
  • the Applicant has also shown that adenoviruses of animal origin are in no way trans-complemented by adenoviruses of human origin, which eliminates any risk of recombination and of propagation in vivo, in the presence of a human adenovirus, which can lead to the formation of an infectious particle.
  • the use of adenoviruses or adenovirus regions of animal origin is therefore particularly advantageous since the risks inherent in the use of viruses as vectors in gene therapy are even lower.
  • the adenoviruses of animal origin which can be used in the context of the present invention can be of canine, bovine, murine origin (example: Mavl, Beard et al., Virology 75 (1990) 81), ovine, porcine, avian or else simienne (example: after-sales service).
  • serotypes 1 to 10 accessible to ATCC such as for example the strains Phelps (ATCC VR-432), Fontes (ATCC VR-280), P7-A (ATCC VR- 827), IBH-2A (ATCC VR-828), J2-A (ATCC VR-829), T8-A (ATCC VR-830), K-ll (ATCC VR-921) or the strains referenced ATCC VR- 831 to 835.
  • bovine adenoviruses it is possible to use the various known serotypes, and in particular those available at ATCC (types 1 to 8) under the references ATCC VR-313, 314, 639-642, 768 and 769. It is possible to use also mention the murine adenoviruses FL (ATCC VR-550) and E20308 (ATCC VR-528), the sheep adenovirus type 5 (ATCC VR-1343), or type 6 (ATCC VR-1340); porcine adenovirus 5359), or simian adenoviruses such as in particular adenovirus referenced to ATCC under the numbers VR-591-594, 941-943, 195-203, etc.
  • adenoviruses or regions of adenoviruses of canine origin are used in the context of the invention, and in particular all the strains of adenoviruses CAV2 [Manhattan strain or A26 / 61 ( ATCC VR-800) for example].
  • Canine adenoviruses have been the subject of numerous structural studies. Thus, complete restriction maps of the CAV1 and CAV2 adenoviruses have been described in the prior art (Spibey et al., J. Gen. Virol.
  • the present invention further relates to a process useful for the preparation of AAV.
  • AAV relates to a process for the preparation of AAV characterized in that it comprises the co-transfection, in the presence of tetracycline or one of its analogs, of a cell line comprising in its genome the expression cassette a transcription activator, with an adenovirus comprising at least one gene of AAV origin under the control of a promoter inducible to tetracycline and either a recombinant virus derived from AAV or a plasmid carrying a transgene between the ITRs of the AAV. It is more preferably an adenovirus comprising, as heterologous viral genes, the rep and cap genes.
  • the method according to the invention takes advantage of the ability to induce the expression of these rep and cap genes placed under the control of a promoter inducible by tetracycline within an adenovirus, in the presence of a sufficient amount of the tetracycline and a transcription activator.
  • this process has the advantage of being simplified in terms of handling compared to a conventional process.
  • a co-infection of a cell line is implemented with an adenovirus as claimed and a recombinant virus derived from an AAV.
  • this method uses a cell line comprising in its genome an expression cassette for the protein known as transcription activator consisting of a first polypeptide capable of binding, in the presence of tetracycline or one of its analogs, to the regulatory sequence of the inducible promoter present in the adenovirus, associated with a second polypeptide which activates transcription.
  • transcription activator As regards more particularly the protein called transcription activator, it is therefore characterized by its ability to bind, in the presence of tetracycline, to the so-called regulatory sequence and its capacity to activate the minimal promoter which is associated with it. As explained above, it is a protein made up of two polypeptides, a first polypeptide which binds to the tet operator sequences, in the presence of tetracycline or an analog thereof, and a second polypeptide whose function is more specifically to activate said transcription.
  • the first polypeptide of the protein known as transcriptional activator is a repressor of tetracycline mutated so as to manifest a behavior opposite to that of a wild repressor, that is to say that it does not binds to operating tet sequences only in the presence and not in the absence of tetracycline.
  • This type of mutation can be carried out according to conventional biological techniques of the mutagenesis type.
  • the difference in amino acids between the wild-type repressor and the mutated repressor according to the present invention can consist of a substitution, deletion and / or addition of one or more amino acids. It has the effect of giving the repressor thus transformed two functional properties: it can bind to the regulatory sequence represented by tetracycline operators by analogy to the wild repressor; however it is inversely regulated by tetracycline.
  • the so-called Tn repressor may be mentioned more particularly. 10 which belongs to class B.
  • the repressor used is derived from this repressor wild TnlO. More specifically, it is a Tn10 repressor mutated into at least one amino acid located in position 71, 95, 101 or 102.
  • the second polypeptide present in the protein can be any already known transcriptional activation domain.
  • it is the activation domain of protein 16 of the herpes simplex virus, more particularly of the 130 amino acids of the C-terminus of VP16 and more preferably of the 11 amino acids of this C terminal end of VP16 or of peptide portions of the C terminal part of VP16 (Sceipel K. et al EMBO J. 1992; 13, 4961-4968) or of derivatives.
  • the expression cassette for this transcription activator is preferably integrated into the genome of a cell line 293.
  • this transcription activator is also placed, in the cell line, under the control of a promoter inducible to tetracycline or one of its analogs as defined above. More preferably, it is a 293 cell line integrating in its genome the cassette Op2 / Tk-TetR-VP16.
  • the present invention also relates to a cell line comprising in its genome an expression cassette for a transcription activator as defined above, whether or not comprising an inducible promoter as defined according to the invention. More preferably, it is a cell line integrating into its genome the O ⁇ 2 / Tk-TetR-VP16 cassette.
  • the invention also relates to the use of this type of cell line for producing adenoviruses according to the invention or AAVs.
  • the present invention also relates to a process for the preparation of adenoviruses comprising at least one of their genes whose expression is under the control of a promoter inducible by tetracycline.
  • the defective recombinant adenoviruses according to the invention can be prepared in different ways.
  • a first method consists in transfecting the DNA of the defective recombinant virus prepared in vitro (either by ligation or in the form of a plasmid) in a competent cell line, that is to say carrying in trans all the functions necessary for complementation virus, and a transcription activator. These functions are preferably integrated into the genome of the cell, which makes it possible to avoid the risks of recombination, and confers increased stability on the cell line.
  • the DNA of the defective recombinant virus carrying the appropriate deletions, one or more viral genes under the control of an inducible promoter tetracycline and one or more therapeutic genes.
  • the deletions are generally carried out on the DNA of the defective recombinant virus, by carrying out digests using appropriate restriction enzymes, then ligations, according to molecular biology techniques, as illustrated in the examples.
  • the viral or therapeutic genes and the inducible promoter can then be inserted into this DNA by enzymatic cleavage then ligation, at the level of the selected regions and in the chosen orientation.
  • the DNA thus obtained which therefore carries the appropriate deletions, one or more viral genes under the control of a promoter inducible to tetracycline and one or more therapeutic genes makes it possible to directly generate the claimed recombinant adenovirus.
  • the DNA of a first recombinant virus carrying the appropriate deletions (or a part of said deletions) and an inducible promoter such as for example Op2 / Tk is constructed, by ligation or in the form of a plasmid. This DNA is then used to generate a first recombinant virus carrying said deletions with an inducible promoter.
  • the DNA of this first virus is then isolated and co-transfected with a second plasmid or the DNA of a defective second recombinant virus carrying the appropriate deletions in particular a deletion in the E1 region, a region allowing homologous recombination and the case if necessary, a therapeutic gene.
  • This second step thus generates the recombinant virus according to the invention.
  • the present invention also relates to any pharmaceutical composition comprising one or more recombinant adenoviruses as described above.
  • compositions of the invention can be formulated for topical, oral, parenteral, intranasal, intravenous, intramuscular, subcutaneous, intraocular, transdermal, etc. administration.
  • the pharmaceutical composition contains pharmaceutically acceptable vehicles for an injectable formulation.
  • pharmaceutically acceptable vehicles for an injectable formulation can be in particular saline solutions (monosodium phosphate, disodium, sodium chloride, potassium, calcium or magnesium, etc., or mixtures of such salts), sterile, isotonic, or dry compositions, in particular lyophilized, which, by addition, as appropriate, of sterilized water or physiological saline, allow the constitution of injectable solutes.
  • the doses of virus used for the injection can be adapted according to various parameters, and in particular according to the mode of administration used, the pathology concerned, the gene to be expressed, or even the duration of the treatment sought.
  • the recombinant adenoviruses according to the invention are formulated and administered in the form of doses of between 10 4 and 10 * 4 pfu / ml, and preferably 10 ⁇ to 10 ⁇ pfu / ml.
  • the term pfu (“plaque forming unit”) corresponds to the infectious power of a virus solution, and is determined by infection of a culture appropriate cell count, and measuring, usually after 5 days, the number of plaques of infected cells. The techniques for determining the pfu titer of a viral solution are well documented in the literature.
  • the adenoviruses of the invention can be used for the treatment or prevention of many pathologies. They are particularly advantageous for the treatment of hyperproliferative pathologies (cancers, restenosis, etc.), by direct injection at the site concerned.
  • the present invention also relates to a method for the destruction of proliferative cells comprising the infection of said cells or of a part of them with an adenoviral vector as defined above.
  • the suicide gene is a gene which confers sensitivity to a therapeutic agent
  • the method of destruction according to the invention then comprises the treatment of the cells with said therapeutic agent.
  • the invention also relates to products comprising a recombinant adenovirus as defined above in which the suicide gene is a gene conferring sensitivity to a therapeutic agent; and said therapeutic agent, as a combination product for simultaneous, separate or spread over time use for the treatment of hyperproliferative pathologies.
  • the suicide gene is a thymidine kinase gene and the therapeutic agent is gancyclovir or acyclovir or an analog.
  • Recombinant vectors according to the invention have particularly attractive properties for use in gene therapy. These vectors indeed combine very high infection, safety and gene transfer properties.
  • Figure 1 Genetic organization of the Ad5 adenovirus. The complete sequence of Ad5 is available on the database and allows those skilled in the art to select or create any restriction site, and thus to isolate any region of the genome.
  • Figure 2 Genetic organization of the E4 region.
  • the plasmids of the pBR322, pUC type and the phages of the Ml 3 series are of commercial origin (Bethesda Research Laboratories).
  • the DNA fragments can be separated according to their size by electrophoresis in agarose or acrylamide gels, extracted with phenol or with a phenol / chloroform mixture, precipitated with ethanol and then incubated in the presence of the DNA ligase from phage T4 (Biolabs) according to the supplier's recommendations.
  • the filling of the protruding 5 ′ ends can be carried out by the Klenow fragment of DNA Polymerase I of E. coli (Biolabs) according to the supplier's specifications.
  • the destruction of the protruding 3 ′ ends is carried out in the presence of the DNA polymerase of phage T4 (Biolabs) used according to the manufacturer's recommendations.
  • the destruction of the protruding 5 ′ ends is carried out by gentle treatment with nuclease SI.
  • Mutagenesis directed in vitro by synthetic oligodeoxynucleotides can be carried out according to the method developed by Taylor et al. [Nucleic Acids Res. 13 (1985) 8749-8764] using the kit distributed by Amersham. Enzymatic amplification of DNA fragments by the so-called PCR technique
  • the verification of the nucleotide sequences can be carried out by the method developed by Sanger et al. [Proc. Natl. Acad. Sci. USA, 74 (1977) 5463-5467] using the kit distributed by Amersham.
  • - Human embryonic kidney line 293 (Graham et al., J. Gen. Virol. 36 (1977) 59). This line contains in particular, integrated into its genome, the left part of the genome of the human adenovirus Ad5 (12%).
  • - Human cell line KB From a human epidermal carcinoma, this line is accessible to the ATCC (ref. CCL17) as well as the conditions allowing its culture.
  • Hela Human cell line Hela: From a carcinoma of the human epithelium, this line is accessible to the ATCC (ref. CCL2) as well as the conditions allowing its culture.
  • MDCK canine cell line The culture conditions for MDCK cells have been described in particular by Macatney et al., Science 44 (1988) 9.
  • DBP6 cell line (Brough et al., Virology 190 (1992) 624). This line consists of Hela cells carrying the E2 gene of adenovirus under the control of the LTR of MMTV.
  • This plasmid carries the sequence of the minimum promoter Tk preceded by two sequences of the tetracycline operator, these sequences are recognized by the tetracycline repressor when it is attached to tetracycline.
  • the plasmid pIC20H (Marsh et al., Gene 32 (1984) 481) is digested with Clal-BamHI and the sequence SEQ ID No. 5, comprising two tetracycline operators upstream of a minimum thymidine kinase promoter, is introduced between these two sites.
  • This plasmid is obtained by ClaI digestion of the plasmid ⁇ IC20H / Op2Tk and insertion of an Hpa2 fragment containing the ITR of Ad5 (coordinates: 1 / + 122).
  • This fragment comes from the commercial vector pSL1180 (Pharmacia) digested with Hind3, site into which the ITR produced by PCR is introduced. with Hind3 sites on each side of the amplified fragment.
  • rTR-Op2-TKprom The following order: rTR-Op2-TKprom.
  • This plasmid corresponds to the plasmid pIC20H / ITR-Op2Tk digested with Hind3, site into which is inserted the Nhe-Xbal fragment of PY6 containing the E4 region of Ad5.
  • plasmid ⁇ PY6 it is obtained according to the following protocol:
  • a plasmid pPY2 is prepared from the plasmid pIC20H.
  • This plasmid pPY2 corresponds to the cloning of the Avr2-SalI fragment (approximately 1.3 kb including the MMTV promoter) of the plasmid pMSG (Pharmacia) between the Xbal and Sali sites of the plasmid pIC20H prepared from an E. coli dam + context.
  • the plasmid pPY4 is derived from the plasmid pPY2 by deletion of a 35 bp fragment after cleavage with BamHI and Bgl2 and then religation.
  • the plasmid pPY5 corresponds to the plasmid pIC20H in which the Taql-Bgl2 fragment including the E4 region of the adenovirus type 5 located between positions 35576 (Taql) and 32490 (Bgl2), was cloned between the ClaI and BamHI sites.
  • the E4 region of the plasmid pPY5 is therefore included in an EcoRV-Sphl fragment which it is cloned after partial digestion between the SmaI and SphI sites of the plasmid pPY4, which generates the plasmid pPY6.
  • the plasmid pIC20H / ITR-Op2Tk-E4-L5 is digested with Xbal and Nrul to recover the corresponding fragment which carries ITR, Op2, the Tk promoter, E4 and L5 in order.
  • This fragment is inserted into the Xbal and Nrul sites of the plasmid pYG4 which contains the entire sequence of the adenovirus from the Xbal site to the Sphl site.
  • This plasmid pYG4-EP is a vector pIC20H in which the Sphl-Xbal fragment of Ad5 (coordinates: 25095-28590) is inserted between its Sphl and Xbal sites.
  • This pYG4-EP vector, deleted from the E3 viral adeno region has sufficient adenoviral sequences between the Sphl and Xbal sites to allow the complementary recombination of the adenovirus for the production of a recombinant adenovirus.
  • This intermediate plasmid makes it possible to introduce an EcoRI site downstream of Op2-Tk.
  • the presence of a restriction site in this position has two advantages. It is used to introduce this promoter upstream of rep-cap after having deleted the p5 promoter and it also makes it possible to insert this hybrid promoter upstream of TetR-VP16 for the preparation of a transformed 293 cell line as described in Example 3 below. -after.
  • the plasmid pIC20H / Op2-Tk obtained according to the protocol described in Example 1, is digested with BamHI, treated with T4 DNA polymerase to make the ends blunt and then redigested by EcoRV, the fragment originating from this digestion and carrying the Op2-Tk promoter is introduced into the EcoRV site of the commercial plasmid pBSSK +.
  • the orientation of the fragment is selected for the presence of an EcoRI site downstream of the promoter.
  • an EcoRI site is introduced at the +1 transcriptional level of the p5 promoter upstream of the coding sequence of Rep78 by the PCR technique on the plasmid ⁇ AV2 (Laughlin C, Gene (1983), 23, 69- 73). This reaction was carried out using the oligonucleotides:
  • This intermediate plasmid makes it possible to join the inducible promoter with rep.
  • the SalI-EcoRI fragments of pXL2630 and EcoRI-NruI of pMA4 are introduced at the Xhol (compatible with SalI) and Nrul sites of pIC20R (Marsh et al., Gene 22 (1984) 481) to give the plasmid pMA6.
  • the fragment thus generated of the plasmid pCA which contains the left end of the genome of the adenovirus Ad5 was then cloned between the EcoRI and Smal sites of the vector pIC20H (Marsh et al., Gene 32 (1984) 481). This generates the plasmid pCB.
  • the plasmid pCB was then cut with EcoRI, its prominent 5 'ends were filled with the klenow fragment of DNA polymerase I from E. coli, then it was cut with BamHI.
  • the fragment thus generated of the plasmid pCB which contains the left end of the genome of the adenovirus Ad5 was then cloned between the Nrul and BglII sites of the vector pIC20H. This generates the plasmid pCE, an interesting characteristic of which is that it has the first 382 base pairs of the adenovirus Ad5 followed by a cloning multisite.
  • the Sau3A (3346) - SstI (3645) fragment and the SstI (3645) - Narl (5519) fragment of the Ad5 adenovirus genome were first ligated and cloned between the ClaI and BamHI sites of the vector pIC20H, this which generates the plasmid pPY53.
  • the Sali- fragment Taql of the plasmid pPY53 prepared from a dam- context, containing the part of the genome of the adenovirus Ad5 between the Sau3A (3346) and Taql (5207) sites was then cloned between the SalI and ClaI sites of the vector pIC20H , which generates the plasmid pCA '.
  • the Narl (5519) - Nrul (6316) fragment of the Ad5 adenovirus genome prepared from a dam context and the Sall-Narl fragment of the pCC plasmid were then ligated and cloned between the SalI and Nrul sites of the vector. pIC20R. This generates the plasmid pCD '.
  • the EcoRV-SnaBI fragment of pMA6 carrying AAV Op2-Tk-rep-cap-polyA + (up to the SnaBI site position 4495 on the AAV sequence) is introduced into the EcoRV site of pCO1 in the two orientations by compared to the adenovirus ITR.
  • the plasmids thus obtained are designated pMA7 (orientation of the cassette in the opposite direction to the ITR of the adenovirus) and pMA8 (same orientation).
  • This section describes the construction of a defective recombinant adenovirus carrying the AAV's Op2-Tk-rep-cap-polyA + cassette.
  • This adenovirus is obtained by co-transfection of the plasmid pMA7 or pMA8 with a deficient adenoviral vector, in helper cells (line 293) providing in trans the functions coded by the El (E1A and E1B) regions of adenovirus.
  • the adenoviruses AdMA7 and AdMA8 were prepared by homologous in vivo recombination between the adenovirus AdRSV ⁇ gal and the plasmids pMA7 and pMA8 according to the following protocol: the plasmid pMA7 or pMA8 linearized by Ndel and the adenovirus AdRSVBgal linearized with Clal are cotransfected in line 293 in the presence of calcium phosphate to allow recombination. The recombinant adenoviruses thus generated are selected by plaque purification.
  • the recombinant adenovirus is amplified in the cell line 293, which leads to a culture supernatant containing the unpurified recombinant defective adenovirus having a titer of approximately 1010 pfu / ml.
  • the viral particles are centrifuged on a cesium chloride gradient according to known techniques (see notably Graham et al., Virology 52 (1973) 456).
  • the AdMA7 or AdMA8 adenovirus is stored at -80 ° C in 20% glycerol.
  • the plasmid pMA28 contains the entire sequence of Ad (E1-, E3-) carrying Op2: Tk repcap polyA + AAV in the E3 region. It was constructed by recombination in E. Coli by introducing the plasmid pMA24 for example in the strain C2110 (pXL2638) (El-, E3-) described in application PCT / FR96 / 00215 included here by reference.
  • the Xbal-Xbal fragment of the pMA7 plsmid carrying Op2: Tk repcap polyA + AAV was introduced at the Xbal site of pYG4-EP in place of the E3 region so that Op2: Tk repcap polyA + AAV is in the reverse orientation that of the E3 region.
  • the palsmide thus constructed is pMA22. 8.2 Construction of the plasmid pMA24 used to carry out the recombination in E.coli:
  • the Nhel-Spel fragment of pMA22 containing the Op2 Tk repcap polyA + AAV cassette flanked by sequences from 27082 to 28593 and from 3471 to 31509 of the Adenovirus was introduced at the compatible site of the plasmid pXL2756 to generate the plasmid pMA24 carrying the regions necessary for the recombination surrounding the Op2 cassette: Tk repcap polyA + AAV, the sacB gene of B. subtilis and the kanamycin resistance gene.
  • This plasmid was then transfected into 293 cells after digestion with Pacl
  • This part describes the construction of a line 293 carrying integrated in its genome the cassette of the hybrid transactivator TetR-VP16 under the control of the promoter Op2-Tk.
  • the plasmid pMA2 was constructed to establish a line by cotransfection of this plasmid pMA2 with a plasmid pMSCV (Hawley et al. J.Exp.Med. (1993), voll76, 1149-1163) carrying the resistance gene neomycin under the control of the promoter PGK (phosphoglyceratekinase).
  • pMA2 is constructed by inserting the SalI-EcoRI fragment from pXL2630 between the XhoI-EcoRI compatible sites of a plasmid pUHD17.1.
  • the plasmid pUHD17.1 is a plasmid comprising the sequences coding for a mutant tetracycline repressor operably linked to the sequence VP16.
  • This vector is derived from the vector pUHD15.1 (H. Bujard; PNASUSA 1992, 89, 55476-5551) which comprises the sequence of the tetracycline repressor W wO w 9 ? 7 / / / 0 ⁇ 0 ⁇ 9v4 ⁇ 7 / PCT / FR96 / 00968
  • the 293 Op2-Tk-TetR-VP16 line of the invention was constructed by co-transfection of the selected cells in the presence of calcium phosphate, by the plasmids pMA2 and pMSCV and a construct coding for the glucocorticoid receptor (Hollenberg and al, 1985). More specifically, the cells of line 293 in dishes 5 cm in diameter were transfected with 1 to 5 ⁇ g of plasmid pMA2.
  • the cells After transfection of the cells, these are washed, then the culture medium (MEM, Sigma) supplemented with fetal wish serum (7% final) is added and the cells are incubated for 20 hours. The following day, the cells are selected in the presence of geneticin G418 (Gibco-BRL, Life Technologies) at the
  • a number of clones were isolated, amplified and selected for their capacity to express a reporter gene, for example lacZ, under the control of the Op2-Tk promoter after addition of an appropriate concentration of tetracycline.
  • the plasmid used is pMA9 and was constructed by the introduction of a StuI-BamHI fragment from pRSVgalIX carrying the sequence coding for E. coli ⁇ -galactosidase and a signal nuclear localization in the plasmid pMA2 previously linearized with EcoRI; treated with bacteriophage T4 DNA polymerase to make its ends blunt and then digested with BamHI.
  • NAME RHONE POULENC RORER S.A.
  • ATCGATACTT TTCTCTATCA CTGATAGGGA GTGGTCTCGA GACTTTTCTC TATCACTGAT 60

Landscapes

  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Plant Pathology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Virology (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
EP96922971A 1995-06-23 1996-06-20 Rekombinante adenovirus,ihre verwendung zur herstellung von aav,zur komplementation falige zellinie und diese enthaltende pharmazeutische zusammensetzungen Withdrawn EP0833896A1 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
FR9507570 1995-06-23
FR9507570A FR2735789B1 (fr) 1995-06-23 1995-06-23 Adenovirus recombinants, leur utilisation pour preparer des aav, lignee cellulaire complementaire et compositions pharmaceutiques les contenant
PCT/FR1996/000968 WO1997000947A1 (fr) 1995-06-23 1996-06-20 Adenovirus recombinants, leur utilisation pour preparer des aav, lignee cellulaire complementaire et compositions pharmaceutiques les contenant

Publications (1)

Publication Number Publication Date
EP0833896A1 true EP0833896A1 (de) 1998-04-08

Family

ID=9480335

Family Applications (1)

Application Number Title Priority Date Filing Date
EP96922971A Withdrawn EP0833896A1 (de) 1995-06-23 1996-06-20 Rekombinante adenovirus,ihre verwendung zur herstellung von aav,zur komplementation falige zellinie und diese enthaltende pharmazeutische zusammensetzungen

Country Status (16)

Country Link
US (3) US6420170B1 (de)
EP (1) EP0833896A1 (de)
JP (1) JP3821846B2 (de)
KR (1) KR100514070B1 (de)
AU (1) AU716508B2 (de)
BR (1) BR9608965A (de)
CA (1) CA2222270A1 (de)
CZ (1) CZ295934B6 (de)
FR (1) FR2735789B1 (de)
HU (1) HU224679B1 (de)
IL (2) IL122710A (de)
MX (1) MX9709549A (de)
NO (1) NO975953L (de)
SK (1) SK174297A3 (de)
WO (1) WO1997000947A1 (de)
ZA (1) ZA965306B (de)

Families Citing this family (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6265212B1 (en) 1995-06-15 2001-07-24 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
DE69638058D1 (de) 1995-06-15 2009-11-26 Crucell Holland Bv Verpackungssysteme für humane rekombinante Adenoviren zur Gentherapie
US6783980B2 (en) 1995-06-15 2004-08-31 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
FR2737222B1 (fr) * 1995-07-24 1997-10-17 Transgene Sa Nouveaux vecteurs viraux et lignee pour la therapie genique
US5891718A (en) * 1996-03-27 1999-04-06 Vical Incorporated Tetracycline inducible/repressible systems
EP0931158A1 (de) * 1996-09-06 1999-07-28 The Trustees Of The University Of Pennsylvania Indutierbare methode zur herstellung von rekombinanten adeno-assoziierten viren mittels t7 polymerase
EP0860445A1 (de) 1997-02-18 1998-08-26 Hoechst Aktiengesellschaft Neue Nukleotidsequenzen für zellzyklusabhängige Expression von Strukturgenen
EP0859008A3 (de) * 1997-02-18 2000-04-05 Hoechst Aktiengesellschaft Nucleinsäurekonstrukt für zellzyklusabhängige Expression von Strukturgene
US6696423B1 (en) 1997-08-29 2004-02-24 Biogen, Inc. Methods and compositions for therapies using genes encoding secreted proteins such as interferon-beta
US6670188B1 (en) 1998-04-24 2003-12-30 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
WO2000016799A1 (en) * 1998-09-23 2000-03-30 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Methods of treating chronic pain
WO2000023592A2 (en) * 1998-10-19 2000-04-27 Powderject Vaccines, Inc. Minimal promoters and uses thereof
CN1074459C (zh) * 1998-11-25 2001-11-07 马丁 重组腺病毒-胸苷激酶构建体及其获得方法和用途
EP1083230A1 (de) * 1999-09-10 2001-03-14 Academisch Medisch Centrum Amsterdam Virale Replikons und von induzierenden Wirkstoffen abhängige Viren
US7115391B1 (en) * 1999-10-01 2006-10-03 Genovo, Inc. Production of recombinant AAV using adenovirus comprising AAV rep/cap genes
GB0027088D0 (en) * 2000-11-06 2000-12-20 Glaxo Group Ltd DNA expression vectors
EP1377672A2 (de) * 2001-02-23 2004-01-07 Novartis AG Vektorkonstrukte
KR100432953B1 (ko) * 2001-09-01 2004-05-28 김주항 개선된 종양 살상 효과를 나타내는 재조합 아데노바이러스
US7569217B2 (en) * 2001-09-24 2009-08-04 University Of Saskatchewan Porcine adenovirus E1 and E4 regions
CA2369985A1 (en) * 2002-01-18 2003-07-18 Duke University Generation of recombinant adeno-associated viral vectors by a complete adenovirus-mediated approach
KR100697245B1 (ko) * 2005-03-28 2007-03-21 한국생명공학연구원 중금속 유도성 프로모터와 이의 생물공학적 활용
WO2013052915A2 (en) 2011-10-05 2013-04-11 Genelux Corporation Method for detecting replication or colonization of a biological therapeutic
US20140087362A1 (en) 2012-03-16 2014-03-27 Aladar A. Szalay Methods for assessing effectiveness and monitoring oncolytic virus treatment
US20130280170A1 (en) 2012-04-20 2013-10-24 Aladar A. Szalay Imaging methods for oncolytic virus therapy
WO2014055960A1 (en) 2012-10-05 2014-04-10 Genelux Corporation Energy absorbing-based diagnostic and therapeutic methods employing nucleic acid molecules encoding chromophore-producing enzymes
WO2015103438A2 (en) 2014-01-02 2015-07-09 Genelux Corporation Oncolytic virus adjunct therapy with agents that increase virus infectivity
CA3126536C (en) 2014-10-14 2023-07-25 Halozyme, Inc. Compositions of adenosine deaminase-2 (ada2), variants thereof and methods of using same
KR20190006495A (ko) 2016-04-15 2019-01-18 알파인 이뮨 사이언시즈, 인코포레이티드 Cd80 변이체 면역조절 단백질 및 그의 용도
KR102536850B1 (ko) 2016-04-15 2023-05-26 알파인 이뮨 사이언시즈, 인코포레이티드 Icos 리간드 변이체 면역조절 단백질 및 그의 용도
US11834490B2 (en) 2016-07-28 2023-12-05 Alpine Immune Sciences, Inc. CD112 variant immunomodulatory proteins and uses thereof
WO2018022946A1 (en) 2016-07-28 2018-02-01 Alpine Immune Sciences, Inc. Cd155 variant immunomodulatory proteins and uses thereof
US11471488B2 (en) 2016-07-28 2022-10-18 Alpine Immune Sciences, Inc. CD155 variant immunomodulatory proteins and uses thereof
TWI780070B (zh) 2016-09-20 2022-10-11 德商百靈佳殷格翰維美迪加股份有限公司 新穎之啟動子
CA3036386A1 (en) 2016-09-20 2018-03-29 Boehringer Ingelheim Vetmedica Gmbh Canine adenovirus vectors
CN110072546B (zh) 2016-09-20 2023-10-31 勃林格殷格翰动物保健有限公司 新的猪流感疫苗
JP6952112B2 (ja) 2016-09-20 2021-10-20 ベーリンガー インゲルハイム フェトメディカ ゲーエムベーハーBoehringer Ingelheim Vetmedica GmbH 新規なehv挿入部位orf70
SG11201903407XA (en) 2016-10-20 2019-05-30 Alpine Immune Sciences Inc Secretable variant immunomodulatory proteins and engineered cell therapy
WO2018148462A1 (en) 2017-02-09 2018-08-16 Indapta Therapeutics, Inc. Engineered natural killer (nk) cells and compositions and methods thereof
KR20240039221A (ko) 2017-03-16 2024-03-26 알파인 이뮨 사이언시즈, 인코포레이티드 Pd-l1 리간드 변이체 면역조절 단백질 및 그의 용도
WO2018170023A1 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Pd-l2 variant immunomodulatory proteins and uses thereof
CA3054068A1 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Cd80 variant immunomodulatory proteins and uses thereof
MA50404A (fr) 2017-10-18 2020-08-26 Alpine Immune Sciences Inc Protéines immunomodulatrices à variants de ligand de icos variant, compositions et méthodes associées
US11505782B2 (en) 2018-06-04 2022-11-22 Calidi Biotherapeutics, Inc. Cell-based vehicles for potentiation of viral therapy
CA3177467A1 (en) 2018-06-04 2019-12-12 Calidi Biotherapeutics, Inc. Cell-based vehicles for potentiation of viral therapy
US20210363219A1 (en) 2018-06-15 2021-11-25 Alpine Immune Sciences, Inc. Pd-1 variant immunomodulatory proteins and uses thereof
US11242528B2 (en) 2018-08-28 2022-02-08 Actym Therapeutics, Inc. Engineered immunostimulatory bacterial strains and uses thereof
EP3876951A1 (de) 2018-11-06 2021-09-15 Calidi Biotherapeutics, Inc. Verbesserte systeme für zellvermittelte onkolytische virustherapie
WO2020107002A2 (en) 2018-11-21 2020-05-28 Indapta Therapeutics, Inc. Methods for expansion of natural killer (nk) cell subset and related compositions and methods
CA3120868A1 (en) 2018-11-30 2020-06-04 Alpine Immune Sciences, Inc. Cd86 variant immunomodulatory proteins and uses thereof
EP3931210A1 (de) 2019-02-27 2022-01-05 Actym Therapeutics, Inc. Zur besiedlung von tumoren, tumorresidenten immunzellen und der mikroumgebung des tumors entwickelte immunstimulatorische bakterien
US12024709B2 (en) 2019-02-27 2024-07-02 Actym Therapeutics, Inc. Immunostimulatory bacteria engineered to colonize tumors, tumor-resident immune cells, and the tumor microenvironment
EP4139439A1 (de) 2020-04-22 2023-03-01 Indapta Therapeutics, Inc. Zusammensetzungen natürlicher killerzellen (nk) und verfahren zur erzeugung davon
US20230203535A1 (en) * 2020-05-26 2023-06-29 St. Jude Children's Research Hospital, Inc. Recombinant P5 Promoter for Use in Reducing DNA Contamination of AAV Preparations
TW202241479A (zh) 2020-12-30 2022-11-01 美商安迅生物製藥公司 遞送外來抗原之溶瘤病毒及表現靶向外來抗原之嵌合受體之經工程化免疫細胞之組合療法

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2145641C (en) * 1992-12-03 2008-05-27 Richard J. Gregory Pseudo-adenovirus vectors
FR2705686B1 (fr) * 1993-05-28 1995-08-18 Transgene Sa Nouveaux adénovirus défectifs et lignées de complémentation correspondantes.
US5814618A (en) * 1993-06-14 1998-09-29 Basf Aktiengesellschaft Methods for regulating gene expression
US5866755A (en) * 1993-06-14 1999-02-02 Basf Aktiengellschaft Animals transgenic for a tetracycline-regulated transcriptional inhibitor
FR2707664B1 (fr) * 1993-07-13 1995-09-29 Centre Nat Rech Scient Vecteurs viraux et utilisation en thérapie génique.
CN1115414C (zh) * 1993-07-13 2003-07-23 罗纳-布朗克罗莱尔股份有限公司 用于基因治疗的病毒载体
US6686200B1 (en) * 1993-08-31 2004-02-03 Uab Research Foundation Methods and compositions for the large scale production of recombinant adeno-associated virus
US5698443A (en) * 1995-06-27 1997-12-16 Calydon, Inc. Tissue specific viral vectors
FR2716682B1 (fr) * 1994-01-28 1996-04-26 Centre Nat Rech Scient Procédé de préparation de virus adéno-associés (AAV) recombinants et utilisations.
FR2716893B1 (fr) * 1994-03-03 1996-04-12 Rhone Poulenc Rorer Sa Virus recombinants, leur préparation et leur utilisation thérapeutique.
US5639661A (en) * 1994-03-23 1997-06-17 The University Of Iowa Research Foundation Genes and proteins for treating cystic fibrosis
US5756283A (en) * 1995-06-05 1998-05-26 The Trustees Of The University Of Pennsylvania Method for improved production of recombinant adeno-associated viruses for gene therapy
US6110744A (en) * 1996-11-13 2000-08-29 Board Of Regents, The University Of Texas System Diminishing viral gene expression by promoter replacement

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9700947A1 *

Also Published As

Publication number Publication date
JPH11507835A (ja) 1999-07-13
US6420170B1 (en) 2002-07-16
BR9608965A (pt) 1999-06-29
KR100514070B1 (ko) 2005-12-21
US7033826B2 (en) 2006-04-25
HUP9900050A3 (en) 2001-08-28
FR2735789A1 (fr) 1996-12-27
SK174297A3 (en) 1998-09-09
NO975953D0 (no) 1997-12-18
NO975953L (no) 1997-12-18
US20030039634A1 (en) 2003-02-27
CA2222270A1 (fr) 1997-01-09
US20020098165A1 (en) 2002-07-25
IL122710A (en) 2006-10-05
FR2735789B1 (fr) 1997-07-25
AU6363996A (en) 1997-01-22
HUP9900050A1 (hu) 1999-04-28
JP3821846B2 (ja) 2006-09-13
ZA965306B (en) 1997-01-24
CZ295934B6 (cs) 2005-12-14
CZ413097A3 (cs) 1998-03-18
AU716508B2 (en) 2000-02-24
KR19990028307A (ko) 1999-04-15
HU224679B1 (hu) 2005-12-28
WO1997000947A1 (fr) 1997-01-09
MX9709549A (es) 1998-03-31
IL122710A0 (en) 1998-08-16

Similar Documents

Publication Publication Date Title
WO1997000947A1 (fr) Adenovirus recombinants, leur utilisation pour preparer des aav, lignee cellulaire complementaire et compositions pharmaceutiques les contenant
EP0667912B1 (de) Defekte adenovirus-vektoren und deren verwendung in der gentherapie
EP0741793B1 (de) Verfahren zur herstellung rekombinanter adeno-associated viren (aav) und deren verwendung
EP0946741B1 (de) Verfahren zur herstellung von rekombinanten viren
WO1996022378A1 (fr) Cellules pour la production d'adenovirus recombinants
EP0787198A1 (de) Defektive adenoviren, die ein theraputisches gen und ein immunschützendes gen enthalten
EP0748385A1 (de) Integriertende rekombinante adenoviren, ihre herstellung und therapeutische verwendungen
FR2707664A1 (fr) Vecteurs viraux et utilisation en thérapie génique.
EP0783585B1 (de) Defektive rekombinante adenoviren mit einem inaktivierten gen iv a 2
EP0946740A1 (de) Neue konstruktionen und vektoren für gezielte und induzierbare gen-expression
FR2718749A1 (fr) Vecteurs viraux et utilisation en thérapie génique.
FR2729674A1 (fr) Cellules pour la production d'adenovirus recombinants
Morsy et al. Helper-dependent adenoviral vectors as gene delivery vehicles
FR2741891A1 (fr) Cellules pour la production d'adenovirus recombinants
FR2738575A1 (fr) Cellules pour la production d'adenovirus recombinants

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19971229

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU NL PT SE

AX Request for extension of the european patent

Free format text: SI PAYMENT 971229

17Q First examination report despatched

Effective date: 20030320

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20070822