EP0918875A1 - Neue interne ribosomenzugangsstelle und diese beinhaltender vektor - Google Patents

Neue interne ribosomenzugangsstelle und diese beinhaltender vektor

Info

Publication number
EP0918875A1
EP0918875A1 EP98922883A EP98922883A EP0918875A1 EP 0918875 A1 EP0918875 A1 EP 0918875A1 EP 98922883 A EP98922883 A EP 98922883A EP 98922883 A EP98922883 A EP 98922883A EP 0918875 A1 EP0918875 A1 EP 0918875A1
Authority
EP
European Patent Office
Prior art keywords
nucleotide
vector
gene
sequence
interest
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Withdrawn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
EP98922883A
Other languages
English (en)
French (fr)
Inventor
Marcelo Lopez Lastra
Caroline Gabus-Darlix
Jean-Luc Darlix
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.)
Institut National de la Sante et de la Recherche Medicale INSERM
Original Assignee
Institut National de la Sante et de la Recherche Medicale INSERM
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 Institut National de la Sante et de la Recherche Medicale INSERM filed Critical Institut National de la Sante et de la Recherche Medicale INSERM
Publication of EP0918875A1 publication Critical patent/EP0918875A1/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Withdrawn legal-status Critical Current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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13051Methods of production or purification of viral material
    • C12N2740/13052Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES
    • C12N2840/206Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES having multiple IRES
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/55Vectors comprising a special translation-regulating system from bacteria

Definitions

  • the present invention relates to the use of a nucleotide sequence derived from the 5 ′ end of genomic RNA or the proviral DNA of a reticuloendotheliosis virus as internal ribosome entry site (IRES) and / or to improve retroviral packaging. More particularly, it relates to expression vectors comprising this sequence and in particular polycistronic vectors allowing the efficient and stable expression of several genes of interest under the dependence of the same promoter.
  • the present invention finds an interesting application in the field of gene therapy vectors.
  • the vectors are obtained by deletion of at least part of the viral genes which are replaced by the genes of therapeutic interest.
  • Such vectors can be propagated in a complementation line which provides in trans the deleted viral functions to generate a viral particle defective for replication but capable of infecting a host cell.
  • retroviral vectors are among the most used, but mention may also be made of vectors derived from adenoviruses, viruses associated with adenoviruses, poxviruses and herpes viruses. This type of vectors, their organization and their mode of infection are widely described in the literature accessible to those skilled in the art.
  • the retroviral genome consists of a linear, single-stranded RNA with positive polarity.
  • the regulation sequences R and TJ5 and U3 and R present at the 5 'and 3' ends respectively, it carries three genes: gag coding for the proteins of the capsid, pol coding for the reverse transcriptase and the integrase and env coding for the proteins of the envelope.
  • the packaging signals located downstream of the U5 sequences up to the start of the coding region of the gag gene, participate in the dimerization and packaging of the viral RNA in the viral particles.
  • the 5 'end of the genome includes a cap (cap) and the 3' end is polyadenylated.
  • Retroviruses can be classified into 4 subfamilies A to D, based on their morphology. Type C includes the majority of retroviruses including the MLV (Murine Leukemia Virus) and MSV (Murine Sarcoma Virus) viruses used in most gene therapy vectors and the REV (Reticuloendotheliosis Virus) viruses from which the nucleotide sequence of the present invention.
  • ribosomes enter the messenger RNA (mRNA) through the cap located at the 5 'end of all eukaryotic mRNAs.
  • the 40S ribosomal subunits move along the RNA until they meet an appropriate AUG codon to start protein synthesis. Generally, initiation takes place at the first AUG codon. But, if this is in an unfavorable context, the 40S subunits continue until a later AUG codon situated in a better translational context (Kozak, 1984, Nucleic Acid Res. 12, 3873-3893; Kozak, 1991, J. Biol. Chem. 266, 19867-19870; Pain, 1996, Eur. J. Biochem. 236, 747-771).
  • Cellular mRNAs having IRES elements have also described have been those coding for the protein BIP (for Immunoglobulin heavy chain binding protein; Macejak and Sarnow, 1991, Nature 353, 90-94), certain growth factors (Teerink et al., 1995, Biochem. Biophy Acts 1264, 403-408; Vagner et al., 1995, Mol. Cell. Biol. 15, 35-44), the translation initiation factor eIF4G (Gan and Rhoads, 1996, J. Biol. Chem. 271, 623-626) and two yeast transcription factors TFIID and HAP4 (lizuka et al., 1994, Mol. Cell. Biol., 14, 7322-7330).
  • protein BIP for Immunoglobulin heavy chain binding protein
  • certain growth factors Teerink et al., 1995, Biochem. Biophy Acts 1264, 403-408; Vagner et al., 1995, Mol. Cell. Biol. 15, 35-44
  • IRES sites have also highlighted in murine retrotransposons of the VL30 type (Berlioz et al., 1995, J. Virol. 69, 6400-6407) and, more recently in the mRNAs coding for the gag precursor of the Friend (FMLV) and Moloney murine leukemia viruses (MoMLV) (Berlioz and Darlix, 1995, J. Virol. 69, 2214-2222; Vagner et al., 1995, J. Biol. Chem. 270, 20376-20383).
  • FMLV gag precursor of the Friend
  • MoMLV Moloney murine leukemia viruses
  • a new internal ribosome entry site has now been found in the 5 'non-coding region of avian reticuloendotheliosis virus (REV) RNA type A (REV-A) and shown to be effective in initiating the translation of coding sequences placed after it in a monocistronic or dicistonic manner.
  • REV avian reticuloendotheliosis virus
  • the IRES site of the present invention is particularly advantageous compared to those already described in the literature. First, it allows a high level of expression of the cistron it controls. In addition and, unexpectedly, it can also, in the context of a retroviral vector, contribute or improve, in association with an appropriate packaging region, the dimerization or packaging functions, allowing an increase in the viral titer . And finally, because of its weak homology with the murine retroviral sequences used in most gene therapy vectors intended for human use, its use considerably reduces the risk of production of viruses competent for replication.
  • the present invention relates to the use of a nucleotide sequence derived from all or part of the 5 'end of the genomic RNA of a type C retrovirus with the exception of the murine leukemia viruses of Friend (FMLV) and Moloney (MoMLV), as internal ribosome entry site (IRES) in a vector and / or to allow or improve the packaging of a retroviral vector.
  • FMLV murine leukemia viruses of Friend
  • MoMLV Moloney
  • IRS internal ribosome entry site
  • nucleotide sequence is meant a sequence composed of ribo (RNA) or deoxyribonucleotides (DNA).
  • RNA ribo
  • DNA deoxyribonucleotides
  • the 5 ′ end of the genomic RNA of a retrovirus corresponds to the 5 ′ quarter of said RNA which extends from the site of initiation of transcription (nucleotide +1) to approximately 2 kb in direction 3 '.
  • retrovirus is widely defined in basic virology works accessible to those skilled in the art and the essential characteristics have been summarized by way of indication above.
  • derivative refers to a sequence having a type C retroviral origin, but which may have undergone at least one modification with respect to the native sequence.
  • the possible modification (s) include the deletion, addition, substitution and / or mutation of one or more nucleotides (nt). Such modifications may have the aim, for example, of increasing the IRES functions, of packaging, or of introducing adequate restriction sites to facilitate the subsequent cloning steps.
  • the term "derivative" also includes the DNA equivalent of genomic RNA in a modified or unmodified form.
  • the term IRES denotes a site capable of promoting the entry of ribosomes into an RNA molecule in a manner independent of the cap.
  • the IRES function can be exercised in any vector or expression cassette.
  • a sequence used in the context of the present invention can also act as an activating element for the packaging of retroviruses or retroviral vectors by promoting the dimerization of two copies of the retroviral genome and / or the packaging of the dimer in the particles. viral.
  • said sequence is capable of exercising an IRES function and of improving the function encapsidation when introduced into an appropriate retroviral vector.
  • a nucleotide sequence as used in the context of the present invention can be isolated from the 5 ′ end of the genomic RNA or of the proviral DNA of a type C retrovirus or of any plasmid of the state of the technique carrying the retroviral fragment of interest. It goes without saying that it can be generated by any technique in use in the field of art, for example by cloning using appropriate probes, by PCR (Polymerase Chain reaction) or by chemical synthesis.
  • said sequence comprises all or part of the region which follows the U3 domain of the 5 'LTR, up to the codon AUG initiator of the gag gene.
  • nucleotides for the purposes of the present invention, it comprises at least 50 nucleotides, advantageously at least 100 nucleotides, preferably at least 200 nucleotides and preferably at least 300 nucleotides included in said 5 ′ end. But, of course, it can extend beyond in the 5 ′ or 3 ′ direction or include additional sequences.
  • said sequence comprises from 100 to 1500 nucleotides and, in particular, from 300 to 800 nucleotides.
  • a type C retrovirus which is more particularly suitable is selected from the REV viruses (Reticuloendotheliosis virus), MSV (Murine sarcoma virus) and in particular that of Moloney (MMSV), MHV (Mus hortulanus virus), MEV (Mouse endogenous retrovirus) , FMOV (FBR murine osteosarcoma virus), AMLV (AKV murine leukemia virus), MEELV (Mouse endogenous ecotropic murine leukemia virus), SFFV (Friend spleen focus-forming virus), RAS V (rat sarcoma virus), FLV (Feline leukemia virus ), FSV (feline sarcoma virus), EFLV (Cat endogenous proviral feline leukemia virus), SSV (Simian sarcoma virus), GALV (Gibbon ape leukemia virus) and
  • a nucleotide sequence in use in the present invention derives from all or part of the 5 'end of the genomic RNA of a reticuloendotheliosis virus (REV).
  • the REV viruses include in particular different subtypes A, B and T as well as the DIAV viruses (Duck infectious anemia virus), SNV (spleen necrosis virus) and CSV (Chick syncytial virus) (see for example Encyclopedia of Virology, 1994, Enrietto , Reticuloendotheliosis viruses, p 1227-1232 Ed. R. Webster and A. Granoff, Académie Press, Hartourt Brace ⁇ Company Publishers).
  • a REV virus which is very particularly suitable is the avian reticuloendotheliosis virus, in particular that of type A (REV-A).
  • nucleotide sequence comprising at least 100 nucleotides and at most 800 nucleotides (nt) of the 5 ′ non-coding end of the REV-A virus and more particularly a substantially homologous or identical nucleotide sequence to all or part of the sequence presented in the sequence identifier SEQ ID NO: 1.
  • substantially homologous refers to a degree of homology greater than 70%, advantageously greater than 80%, preferably greater than 90% and, most preferably, greater than 95%.
  • said nucleotide sequence may have a sequence slightly different from that described in SEQ ID NO: 1 or 2, provided, however, that the modification or modifications does not affect its IRES functions and / or packaging.
  • the nucleotide sequence used in the context of the present invention is identical to the sequence presented in the sequence identifier SEQ ID NO: 2: (i) starting at nucleotide 1 and ending at nucleotide 578, (ii) starting at nucleotide 265 and ending at nucleotide 578, or
  • the IRES function of said nucleotide sequence is particularly advantageous in a context poor in magnesium ion, for example in a cellular context.
  • a high concentration of Mg 2+ ions can decrease the efficiency of sequence-mediated translation initiation.
  • a nucleotide sequence in use in the present invention is more particularly intended to be integrated into a vector for the transfer and expression of one or more gene (s) of interest.
  • the choice of such a vector is wide and the techniques of cloning into the selected vector are within the reach of those skilled in the art.
  • adenovirus adenovirus
  • baculovirus herpes virus
  • virus associated with an adenovirus or retrovirus Such vectors are widely described in the literature.
  • adenoviral vector it can be derived from a human (preferably type 2 or 5), animal (preferably canine or bovine) adenovirus or a hybrid between various species.
  • the general technology for adenoviruses is disclosed in Graham and Prevec (1991,
  • said nucleotide sequence is preferably positioned upstream of a gene of interest to improve the translation of the expression product for which it codes.
  • It can be implemented in an expression cassette of the monocistronic type (for the expression of a gene of interest placed under the control of a promoter) or polycistronic (for the expression of at least two genes d 'interest placed under the control of the same promoter).
  • the latter can contain several elements in tandem "IRES site-gene of interest" of which at least one of the IRES sites consists of a nucleotide sequence as defined above.
  • Particularly preferred is the use in a dicistronic cassette either upstream of the first gene of interest or upstream of the second, the latter variant being the preferred.
  • a vector according to the invention comprises several expression cassettes, these can be inserted in any orientation relative to each other, either in the same orientation (promoter acting in the same direction) or in reverse orientation ( promoter acting in an opposite orientation).
  • a vector according to the invention can comprise several nucleotide sequences in use according to the invention. In this case, it is preferable that they are derived from different type C retroviruses.
  • a vector according to the invention derives from a retrovirus.
  • retroviruses such as avian erythroblastosis virus (AEV), avian leukemia virus (AVL), avian sarcoma virus (ASV), necrosis virus of spleen (SNV) and Rous sarcoma virus (RSV), bovine retroviruses, feline retroviruses (FLV, FSV ....), murine retroviruses such as murine leukemia virus (MuLV), the virus Friend's (FMLV) and murine sarcoma virus (MSV) and primate retroviruses (GALV, FSV, BAEV ).
  • AEV avian erythroblastosis virus
  • AEV avian leukemia virus
  • ASV avian sarcoma virus
  • SNV necrosis virus of spleen
  • RSV Rous sarcoma virus
  • bovine retroviruses bovine retroviruses
  • retroviral vectors which can be envisaged for the purposes of the present invention comprise at least the following elements which are operatively associated: a 5 'LTR and a 3' LTR retroviral, one or more gene (s) of interest, and the nucleotide sequence use in the context of the present invention to allow or improve the packaging of said vector in a viral particle and / or as an IRES site to allow or promote the expression of a gene of interest positioned downstream of said nucleotide sequence.
  • the retroviral 5 'LTR can be used as a promoter, but an internal promoter can also be used.
  • the 5 ′ and possibly 3 ′ LTR may have the same retroviral origin (for example REV) as the nucleotide sequence or a different origin.
  • a monocistronic vector will comprise from 5 'to 3' a 5 'LTR, the nucleotide sequence, a gene of interest and a 3' LTR.
  • a retroviral vector according to the invention can also comprise a conventional (E +) packaging region.
  • E + conventional packaging region
  • the presence of the latter is not required when the nucleotide sequence in use in the present invention can alone exercise the packaging function.
  • the retroviral LTR 5 ′ derives from a REV virus and, preferably from SNV, and the nucleotide sequence is substantially homologous or identical to the sequence presented in SEQ ID NO: 2, starting at nt 1 and ending at nt 578 or starting at nt 265 and ending at nt 578.
  • a retroviral vector according to the invention comprises at least: (a) a 5 'retroviral LTR,
  • the retroviral vector according to the invention comprises an expression cassette directed by an internal promoter region
  • Other elements can also be included, for example another IRES site and another gene of interest or another expression cassette.
  • a preferred retroviral vector according to the invention comprises an encapsidation region deriving from a murine retrovirus, in particular from a MoMLV, or from a VL30 type retrotransposon and an IRES site comprising a nucleotide sequence substantially homologous or identical to the sequence presented in the sequence identifier SEQ ID NO: 2:
  • the packaging region derives from a MoMLV and the IRES site consists of a nucleotide sequence identical to the sequence presented in SEQ ID NO: 2 starting at nucleotide 265 and ending at nucleotide 578 or starting at nucleotide 452 and ending at nucleotide 578.
  • the packaging region derives from a MoMLV
  • the IRES site consists of a nucleotide sequence identical to the sequence presented in SEQ ID NO: 2 starting at nucleotide 265 and ending at nucleotide
  • a gene of interest in use in the invention can be obtained from a eukaryotic or prokaryotic organism or from a virus by any conventional molecular biology technique. It can code for a polypeptide corresponding to a native protein as found in nature homologous to the host cell or not, a protein fragment, a chimeric protein originating from the fusion of polypeptides of various origins or a mutant with improved and / or modified biological properties. Such a mutant can be generated by substitution, deletion and / or addition of one or more amino acid residues.
  • polypeptide can be (i) intracellular (ii) membrane present on the surface of the host cell or else (iii) secreted outside the host cell and therefore comprise appropriate additional elements, such as a sequence coding for a signal of secretion or a transmembrane anchoring region.
  • a vector according to the invention is particularly intended for the prevention or treatment of cystic fibrosis, hemophilia A or B, Duchenne or Becker's myopathy, cancer, AIDS, cardiovascular diseases (restenosis, arteriosclerosis, ischemia %) and other infectious diseases due to a pathogenic organism: virus, bacteria, parasite or prion.
  • genes of interest which can be used in the present invention are those which code for the following proteins: a cytokine and in particular an interleukin (IL-2, IL-7, IL-10, IL-12 ...), an interferon, a tissue necrosis factor and a growth factor and in particular hematopoietic (G-CSF, GM-CSF), a factor or cofactor involved in coagulation and in particular factor VIII, factor IX, von Willebrand factor, antithrombin III , protein C, thrombin and hirudin, an enzyme and in particular trypsin, a ribonuclease, alkaline phosphatase (plap) and ⁇ -galactosidase, an enzyme inhibitor such as ⁇ l-antitrypsin and inhibitors of viral proteases an expression product of a suicide gene such as thymidine kinase of the HSV virus (herpes virus) type 1, that of the fiirl gene and / or fcy
  • adenosine diaminose glucocerebrosidase and phenylhydroxylase
  • a protein capable of inhibiting the initiation or progression of cancers such as the expression products of tumor suppressor genes, for example the p53, p73 and Rb genes, a protein capable of stimulating an immune response, an antibody, the antigens of the major histocompatibility complex or an immunotoxin
  • a protein capable of inhibiting a viral infection or its development for example the antigenic epitopes of the virus in question or altered variants of viral proteins likely to compete with native viral proteins, a cellular or nuclear receptor or one of their ligands, a growth factor (FGF for Fibroblast Growth Factor, VEGF for Vascular Endothelial cell Growth Factor ...), and a apoptosis inducer (Bax %), an apoptosis inhibitor (Bcl2, BclX %), a cytostatic agent (p21, pl6, Rb %), a n
  • a use in gene of interest in the present invention may also encode a selectable marker to select or identify the transfected host cell with a vector according to the invention.
  • neo gene neomycin
  • dhjr gene dihydrofolate reductase
  • CAT gene Chloramphenicol Acetyl Transferase
  • gpt gene xanthine phosphoribosyl
  • a functional promoter in the host cell considered and, preferably, a human cell.
  • the choice of promoter is very wide and within the reach of those skilled in the art. It may be a promoter naturally governing the expression of a gene of interest in use in the present invention or any other promoter of any origin. Furthermore, it can be constitutive or regulable in nature, in particular in response to certain tissue-specific or event-specific cellular signals. For example, it may be advantageous to target the expression of the gene of interest at the level of lymphocyte cells in the context of AIDS, of lung cells in the context of cystic fibrosis or of muscle cells in the context of myopathies.
  • the promoters suitable for the present invention can be chosen from promoters SV40 (Simian 40 virus), CMV (Cytomegalovirus), HMG (Hydroxymethyl-Glutaryl Coenzyme A), TK (Thymidine kinase), Retroviral LTRs such as that of MoMLV, RSV or MSV when a retroviral vector, the adenoviral El A and late MLP (Major Late Promotor) promoters are used, in particular in the context of an adenoviral vector, the 7.5K promoters, H5R, pKlL, p28 and pi 1 intended for poxviral vectors like the vaccinia virus, the promoter PGK (Phosphoglycero kinase), the liver-specific promoters of the genes coding for the alpha-antitrypsin, factor IX, albumin and transferrin, the promoters of the immunoglobulin genes which allow expression in lymphocytes,
  • Muc-1 gene promoters overexpressed in breast and prostate cancers can be mentioned in particular (Chen et al., 1995, J. Clin. Invest. 96, 2775-2782), CEA (for carcinoma embryonic antigen) overexpressed in colon cancer (Schrewe et al., 1990, Mol. Cell. Biol. 10, 2738-2748), tyrosinase overexpressed in melanomas (Vile et al ., 1993, Cancer Res.
  • ERB-2 overexpressed in breast and pancreatic cancers (Harris et al., 1994, Gene Therapy 1, 170-175), ⁇ -fetoprotein overexpressed in cancers liver (Kanai et al., 1997, Cancer Res. 57, 461-465), APC overexpressed in colorectal cancers, BRCA-1 and 2 (Wooster et al., 1995, Nature 378, 789-792) overexpressed in ovarian cancer and PSA (for prostate specifies antigen) overexpressed in prostate cancer.
  • the gene of interest in use in the present invention may comprise other sequences improving its expression, both at the level of transcription and of translation; for example an enhancer-type transcription activating sequence, an intronic sequence, a transcription termination signal (polyA) and, as indicated above, a secretion signal or a transmembrane region.
  • an enhancer-type transcription activating sequence for example an enhancingr-type transcription activating sequence, an intronic sequence, a transcription termination signal (polyA) and, as indicated above, a secretion signal or a transmembrane region.
  • polyA transcription termination signal
  • the invention also covers the viral particles generated from a viral vector according to the invention. This is generally done by transfection of the latter into a suitable cell line. If the viral vector used is defective for replication, a complementation line will be used. In general, a person skilled in the art knows the lines that can be used to generate infectious viral particles as well as the process to be implemented according to the vector used.
  • adenoviral vector use may be made of line 293 (Graham et al., 1977, J. Gen. Virol., 36, 59-72). Being a retroviral vector, one can consider using ecotropic cell lines, such as the CRE line (Danos and Mulligan, 1988, Proc. Natl. Acad. Sci. USA, 85, 6460-6464) or GP + E-86 (Markowitz et al., 1988, J. Virol., 62, 1120-1124). However, it is particularly preferred to use an amphotropic complementation line such as the PG13 line (Miller et al., 1991, J.
  • infectious viral particles are recovered in the culture supernatant of the transfected complementation cells.
  • the invention also extends to cells comprising a vector according to the invention or infected with infectious viral particles according to the invention.
  • the transfection methods are well known to those skilled in the art. Mention may be made of the calcium phosphate precipitation technique, that of DEAE dextran, microinjection or encapsulation in lipid vehicles.
  • the vectors according to the invention can be present in the host cell in a form integrated into the cell genome or in the form of episomes both in the nucleus and in the cytoplasm.
  • the cell according to the invention is advantageously a eukaryotic cell, in particular a mammalian cell and, preferably, a human cell.
  • It can be a primary or tumor cell of hematopoietic origin (totipotent stem cell, leukocyte, lymphocyte, monocyte, macrophage ...), hepatic, epithelial, fibroblast, of the central nervous system and, in particular, d '' a muscle cell (myoblast, myocyte, satellite cell, smooth muscle ...), cardiac, vascular, tracheal, pulmonary or central nervous system.
  • totipotent stem cell leukocyte, lymphocyte, monocyte, macrophage
  • hepatic epithelial
  • fibroblast of the central nervous system
  • d '' a muscle cell myoblast, myocyte, satellite cell, smooth muscle
  • cardiac vascular, tracheal, pulmonary or central nervous system.
  • the present invention also relates to the therapeutic use of a vector, a viral particle or a cell according to the invention, for the preparation of a pharmaceutical composition intended for the treatment and / or prevention of a disease.
  • treatable by gene therapy including a genetic disease, an acquired disease such as cancer or an infectious disease.
  • a vector according to the invention can be used for other purposes such as the recombinant production in prokaryotic or eukaryotic cells of expression product (s) encoded by at least one of the genes of interest.
  • expression product s
  • the coexpression of an antibiotic resistance gene as a second cistron can make it possible to increase the expression of a first cistron.
  • mice can be mice, rats, rabbits, fish, primates or farm animals (cattle, sheep, pigs ”).
  • the techniques for generating these transgenic animals are known.
  • the polypeptide of interest can be recovered in a conventional manner, for example in the biological fluids (blood, milk, etc.) of the animal.
  • the invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising, as therapeutic or prophylactic agent, a vector, a viral particle or a cell according to the invention or a polypeptide of interest obtained in accordance with the use according to the invention, in combination with a pharmaceutically acceptable vehicle.
  • a pharmaceutical composition according to the invention can be manufactured in a conventional manner.
  • a therapeutically effective amount of such an agent is combined with an acceptable carrier, diluent or adjuvant. It can be administered according to any route of administration and this in a single or repeated dose after a certain time interval.
  • intravenous, intramuscular, intrapulmonary (possibly aerosolized) or intratumoral administration is preferred.
  • the amount to be administered will be chosen according to different criteria, in particular the use as treatment or vaccine, the route of administration, the patient, the type of disease to be treated and its state of evolution, the duration of treatment, the vector retained ... etc.
  • a pharmaceutical composition according to the invention comprises between 10 4 and 10 14 pfu (unit forming plaques), advantageously between 10 5 and 10 13 pfu and, preferably, between 10 6 and 10 "pfu of viral particles.
  • a vector-based composition can be formulated as doses comprising from 0.01 to 100 mg of DNA, preferably from 0.05 to 10 mg and, most preferably, from 0.1 to 5
  • the formulation can also include, alone or in combination, a pharmaceutically acceptable diluent, adjuvant or excipient, as well as a solubilizing, stabilizing or preserving agent. single or in multiple doses in liquid or dry form (lyophilisate ...) capable of being reconstituted extamporanally with an appropriate diluent.
  • the invention relates to a method of treatment of genetic diseases, cancers and infectious diseases according to which a therapeutically effective amount of a vector, a viral particle or a cell according to the invention is administered to a patient having need such treatment.
  • a therapeutically effective amount of a vector, a viral particle or a cell according to the invention is administered to a patient having need such treatment.
  • they can be administered directly in vivo, for example by intravenous, intramuscular, intratumoral injection or by aerosolization in the lungs.
  • an ex vivo gene therapy protocol which consists in removing the cells from a patient (stem cells from the bone marrow, peripheral blood lymphocytes, etc.), transfecting them with a vector according to the invention and cultivating them in vitro before re-implanting them in the patient.
  • the invention relates to the use of a vector, a viral particle or a pharmaceutical composition according to the invention for the transfection or infection of pluripotent cells, in particular pluripotent cells of the central nervous system.
  • Figure 1 is a schematic representation of the monocistronic plasmids used as templates for the in vitro synthesis of capped and non-capped RNAs. They contain the early cytomegalovirus (Po CMV) promoter usable for expression in vivo, the promoter of the gene coding for phage T7 RNA polymerase (Po T7) usable for in vitro experiments, different portions of the 5 end '' untranslated (leader) of the REV-A virus (1 to 578 for pREV CB-95, 578 to 1 for pREV CG-53, 1 to 578 deleted from nt 268 to 452 for pREV CG-54, 265 to 578 for pREV CG-55 and 452 to 578 for pREV CG-56) and the LacZ gene ( ⁇ LacZ) coding for a truncated ⁇ -galactosidase at the C-terminal end with a
  • FIG. 2 is a schematic representation of the dicistronic plasmids used as templates for the in vitro synthesis of capped and uncapped RNA. They contain the early cytomegalovirus promoter (Po CMV) usable for expression in vivo, the promoter of the gene coding for phage T7 RNA polymerase (Po T7) usable for in vitro experiments, the neo gene, different portions of the 5 'untranslated end (leader) of the REV-A virus (1 to 578 for pREV CB-54, 578 to 1 for pREV CG-50, 1 to 578 deleted from nt 268 to 452 for pREV CG-52, 265 to 578 for pREV CB-55 and 452 to 578 for pREV CG-58) and the LacZ gene ( ⁇ LacZ) coding for a truncated ⁇ -galactosidase at the C-terminal end with a molecular mass of approximately
  • Figure 3A is a schematic representation of the dicistronic retroviral vectors having two elements of different retroviral origin, as IRES and packaging region (E) and two genes of interest such as the reporter genes coding for phosphatase alkaline placenta and neo coding for neomycin phosphotransferase.
  • VL30E + corresponds to the 5 'untranslated region of HaMSV and MoMLV E + corresponds to the packaging region of MoMLV.
  • FIG. 4 illustrates the effect of rapamycin on the activities A) alkaline phosphatase and B) neomycin phosphotransferase produced in GP + E-86 cells not transfected or stably transfected by the different vectors pREV HW or pEMCV-CBTV (pCBlOO ) and treated with rapamycin (full boxes) or not treated (control, dotted boxes).
  • Figure 5 illustrates the optimization of the transduction protocol applied to Dev neuroectodermal cells. The percentage of Dev cells transduced by the pEMCV-CBTV (IRES EMCV) and pREV HW-3 viruses (IRES REV-A) is determined by flow cytometry.
  • EXAMPLE 1 Identification of an IRES site at the 5 ′ end of the REV-A RNA.
  • the DNA fragments corresponding to sequences 1 to 578, 265 to 578 and 452 to 578 of the REV-A RNA are isolated by PCR from the pREVSC-1 template (Darlix et al., 1992, J. Virol. 66, 7245-7252). Appropriate primers are used that a person skilled in the art can design, provided at their ends with a Mzel site. After digestion with this enzyme, the PCR fragments are inserted upstream of the LacZ gene in the vector pEMCV-M260-837 (Berlioz et al., 1995, J. Virol. 69, 6400-6407) previously cleaved by Nhel.
  • the LacZ gene used codes for a truncated ⁇ -galactosidase product at the C-terminal end.
  • the monocistronic plasmids pREV CB-95 (1-578), pREV CG-55 (265-578) and pREV CG-56 (452-578) are obtained, illustrated in Figure 1.
  • the dicistronic plasmids pREV CB-54 (1- 578), pREV CB-55 (265-578) and pREV CG-58 (452-578) are shown in Figure 2 and result from the insertion of the previous PCR fragments between the neo and LacZ genes of pEMCV-D260-837 (pCBlOl) (Berlioz et al., 1995, J. Virol. 69, 6400-6407) also subjected to digestion with Nhel.
  • Amplification of nt 1 to 578 deleted from sequences 268 to 452 is carried out using the vector pREVSC-1 previously digested with Kpnl and S ⁇ , treated with the Klenow fragment of AD de polymerase from E. coli and religious.
  • the amplified fragment digested by Mzel is cloned in pEMCV-M260-837 upstream of the LacZ gene or between the neo and LacZ genes of pEMCV-D260-837, the two vectors having been digested with Mzel, to give respectively pREV CG-54 (Fig 1) and pREV CG-52 (Fig 2).
  • monistristrum control plasmids pREV CG-53 (Fig 1) and dicistronic pREV CG-50 (Fig 2) were constructed by introduction of the PCR fragment carrying the sequences REV-A l to 578 in the previous vectors in reverse orientation (578- 1).
  • the initiation of the translation of ⁇ -galactosidase is under the control of the AUG codon of the gag gene of REV-A located at position 574-576, while in the control plasmids, the synthesis of ⁇ -galactosidase depends on an AUG placed in a favorable Kozak context introduced by PCR.
  • RNAs are synthesized from 1 ⁇ g of plasmid DNA linearized by Sspl (position 1240 in the LacZ gene) using T7 RNA polymerase (mMessage mMachine or MAXIscript, Ambion) in a reaction volume 20 ⁇ l according to the protocol indicated by the supplier. Transcription is stopped by treatment of the DNA matrix with the enzyme DNasel followed by precipitation of the RNAs in the presence of lithium chloride. The RNAs are taken up in 50 ⁇ l of TE buffer (10 mM Tris-HCl pH7.5, EDTA ImM) before being purified and desalted by passage through an S-300 MicroSpin TM column (Pharmacia BioTech) according to the supplier's instructions.
  • TE buffer 10 mM Tris-HCl pH7.5, EDTA ImM
  • RNA luciferase is tested in same reaction conditions (positive control).
  • the samples are heat denatured in 62.5 mM Tris-HCl pH 6.8, 2% sodium dodecyl sulphate (SDS), 10% glycerol, 5% ⁇ -mercaptoethanol and 0.02% bromophenol blue and the labeled proteins analyzed by electrophoresis on polyacrylamide gel 12% (weight / vol), 0.2% SDS.
  • SDS sodium dodecyl sulphate
  • the neo gene product and ⁇ -galactosidase migrate at a molecular mass of approximately 28 and 46 kDa respectively.
  • the efficiency of the cap-dependent and independent translation is quantified by CT scan (Phospho-Image ⁇ r Storm 840, version 4.00, Molecular Dynamics; Image Quant TM version 1.1, Molecular Dynamics).
  • the intensity of the labeling of the expression product of the second cistron ( ⁇ -galactosidase) whose translation is mediated by PIRES is evaluated after standardization of the level of expression of the neo product.
  • RNAs obtained from the monocistronic plasmids pREV CB-95, pREV CG-53, pREV CG-54, pREV CG-55 and pREV CG-56 is as efficient as that of the capped RNAs.
  • the amount of ⁇ -galactosidase generated from the plasmid pREV CG-53 in which the REV-A sequences (1 to 578) are in antisense orientation is much lower than that obtained with the constructions using a REV sequence -A in sense orientation.
  • Table 1 Report of gene expression in the presence and absence of FMDV protease L.
  • FIG. 3 illustrates the vectors of the pREV HW series having LTRs of MoMLV type and the control vectors used in the experiments described below. Although these are not represented, vectors designated pMC have also been constructed which differ from pREV HW only in that their LTRs are of SNV origin and negative controls in which the REV-A sequences are positioned in orientation reverse (3 '-> 5') compared to LTRs. For all the molecular biology stages, these vectors are introduced into the plasmid pBR322.
  • the control vector pEMCV-CBTV (pBC100) is a dicistronic vector comprising, in addition to the LTRs and the packaging region derived from MoMLV, the gene coding for placental alkaline phosphatase (plap), the translation of which is cap-dependent and the gene neo. whose translation is site dependent
  • pREV HW-1 the REV-A fragment extending from nt 265 to 578 generated by PCR and digested with zel is cloned between the plap and neo genes of pMLV-CB71 (Berlioz and Darlix, 1995, J. Virol. 69, 2214-2222).
  • pREV HW-2 the EcoRI fragment of pVL CBT5 (Torrent et al., 1996, Human Gene Therapy 7, 603-611) carrying the LTR 5 ′ MoMLV and the packaging sequences of VL30 is introduced into the vector pR ⁇ V HW- 1 linearized by EcoRI.
  • pR ⁇ V HW-3 the EcoRI fragment from p ⁇ MCV-CBTV containing the 5 'LTR and the packaging sequences of MoMLV is inserted into the vector pR ⁇ V HW-1 linearized by EcoRI.
  • pR ⁇ V HW-4 the R ⁇ V-A fragment extending from nt 452 to 578 generated by PCR and digested with Nftel is cloned between the plap and neo genes of pMLV-CB71.
  • pR ⁇ V HW-5 the EcoRI fragment of pVL CBT5 carrying the LTR 5 ′ MoMLV and the packaging sequences of VL30 is introduced into the vector pR ⁇ V HW-4 linearized by EcoRI.
  • pR ⁇ V HW-6 the EcoRI fragment from p ⁇ MCV-CBTV containing the 5 'LTR and the MoMLV packaging sequences is inserted into the vector pR ⁇ V HW-4 linearized by EcoRI.
  • the pMC series vectors are obtained according to the following construction scheme:
  • the S ⁇ V LTRs are generated by PCR from the plasmid R ⁇ V-A 2-20-6 (O'Rear and Temin, 1982, Proc. ⁇ atl. Acad. Sci USA 79, 1230-1234; Darlix et al., 1992, J. Virol. 66, 7245-7252).
  • the neo gene is isolated from pMLV-CB71 by digestion with Sali and BamHI then introduced between the same sites of the vector pUC19 (Gibco BRL).
  • the 5 'LTR of S ⁇ V (nt 1 to 861) is digested with H di ⁇ and Sali, and inserted into pUC19-neo previously cleaved by these same enzymes.
  • the LTR 3 'S ⁇ V (nt 7230-8300) digested with Sm ⁇ l and EcoRI is cloned in the preceding vector to give pCG-61 containing LTR 5' S ⁇ V-neo-LTR 3 'S ⁇ V.
  • a vector designated pCG-62 is generated which differs from the previous one by deleting the env sequences (nt 7230-7691) obtained by BglR-AvrTl, Klenow treatment and religation.
  • the plap gene isolated from the clone Cla-12AP (DGoff) is introduced between the EcoRI and Xbal sites of a bluescript plasmid previously deleted from the Sali site (EcoRI-.X7zoI digestion) before being re-isolated in the form of a Kpnl-SaR fragment and cloned between the same pCG-61 sites and pCG-62, to give pCG-63 and pCG-64 respectively.
  • the LacZ gene is obtained by partial digestion of pR ⁇ V CB-95 with the enzymes Sali and Bam ⁇ l. Its insertion between the SalI and BamUl sites of pCG-61 and pCG-62 gives rise to pCG-65 and pCG-66 respectively.
  • LTR-Gene-LTR block is isolated from each plasmid pCG-62, pCG-64 and pCG-66 by H dlII-EcoRI digestion to be inserted into the vector pBR322 cleaved by these same enzymes. PMCl, pMC2 and pMC3 are generated.
  • the GP + ⁇ -86 ecotropic complementation line (Markowitz et al., 1988, J. Virol., 62, 1120-1124) and the NI ⁇ 3T3 target cells (mouse fibroblastic cells) available at TATCC, are cultured at 37 ° C. in the presence of 5% CO 2 in DM ⁇ M medium (Dulbecco's Modified Eagle's Medium, Gibco BRL) supplemented with 10% newborn calf serum.
  • DM ⁇ M medium Dulbecco's Modified Eagle's Medium, Gibco BRL
  • GP + ⁇ -86 helper cells and NIH3T3 target cells are cultured the day before transfection and infection. Viral infections are carried out according to the conventional protocol described in the literature.
  • the viral supernatants were filtered (on 0.45 ⁇ m filters) and placed in the presence of polybrene to a final concentration of 8 ⁇ g / ml.
  • the infection is continued overnight at 37 ° C. and the following day, the cells are washed and cultured in fresh medium. After 48 h, the cells are placed in a selective medium (1 mg / ml of G418) or stained to determine the number of cells expressing the alkaline phosphatase plap.
  • fixing is carried out in PBSxl buffer containing 2% formaldehyde and 0.2% glutaraldehyde.
  • the cells are washed twice in AP buffer (0.1 M Tris-HCl pH 9.5, 0.1 M NaCl, 50 mM MgCl 2 in PBSxl) and placed for 5 h in the staining solution (0.1 mg / ml of 5-bromo-4-chloro-3-indolyl phosphate (BCIP), 1 mg / ml of a terazolium salt of Nitroblue (NBT) and 1 mM Levamisol in buffer AP).
  • AP buffer 0.1 M Tris-HCl pH 9.5, 0.1 M NaCl, 50 mM MgCl 2 in PBSxl
  • the staining solution 0.1 mg / ml of 5-bromo-4-chloro-3-indolyl phosphate (BCIP), 1 mg / ml of a terazolium salt of Nitroblue (NBT) and 1 mM Levamisol in buffer AP.
  • the titer of the recombinant viruses is determined after transfection of the GP + E-86 ecotropic cells. After two days of incubation, the viral supernatant is harvested and used to determine the viral titer (transient expression). Then, the transfected cells are selected with G418 for one month. After this selection, the viral titer is determined on the harvested supernatant (stable expression). This corresponds to the number of infectious particles per ml of supernatant.
  • the pREV HW-1 and pREV HW-4 vectors lacking a conventional packaging region are incapable of producing infectious viral particles after transfection into the MLV helper line (GP + E-86).
  • the vector pMC1 can be packaged in SNV viral particles after transfection of the helper line SNV D17-C3A2 (for example ATCC CRL8468), indicating that the REV-A sequences extending from nt 265 to 578 can be used in this context as an encapsidation region.
  • Retroviral vectors comprising both a REV-A sequence (265-578 or 452-578) and a conventional packaging region produce high titer viral particles (pREV HW-2, 3, 5 and 6).
  • the association with the packaging region of MLV proves to be particularly advantageous since it gives viral titers 2 (pREV HW-6) to 5 times (pREV HW-3) higher than the reference vector pEMCV-CBTV combining this same encapsidation region and TIRES EMCV.
  • the comparison of the data obtained with the identical vectors varying only at the level of the REV-A segment used suggests that the sequence ranging from nt 265 to 578 is capable of cooperating with the packaging region and thus improving the packaging of viral RNAs and consequently the viral titers.
  • An element interacting positively with packaging could be present between nt 452 and 265 in the REV-A genome.
  • the morphology of the recombinant pREV HW virions produced after transfection of the GP + E-86 line with the corresponding vectors is analyzed by electron microcopying.
  • the viruses obtained from pEMCV-CBTV under the same conditions are used as controls and the wild retroviruses obtained after infection of the NIH3T3 cells with the FMLV-29 strain (Friend murine leukemia virus strain 29). Microscopy results indicate that the RNA content does not affect the morphology of the recombinant viruses.
  • GP + E-86 cells are stably transfected with 20 ⁇ g of vectors of the pREV HW or pEMCV-CBTV series and cultured under selective conditions (G418) for 15 days. At 70 to 80% confluence, they are brought into the presence of rapamycin at a final concentration of 20 ng / ml. The latter has an inhibitory effect on cap-dependent translation varying from 15 to 40% depending on the cell line (Beretta et al., 1996, EMBO J. 15, 658-664) but does not affect cap-independent translation. Cell extracts are conventionally prepared after 20 h of incubation.
  • the cells are washed twice in PBSxl, placed in 1 ml of TEN (40 mM Tris-HCl pH7.5, 1 mM EDTA, 50 mM NaCl) for a 10 cm dish then recovered by scraping and centrifuged on low speed.
  • the pellet is taken up in 100 ⁇ l of 0.25 M Tris-HCl, pH8 and subjected to cell lysis by 3 freeze-thaw cycles. After centrifugation for 10 min at 14,000 g, the supernatant is recovered and can be stored at -70 ° C while waiting for the enzymatic tests.
  • the final protein concentration is determined by the Micro BCA test (Pierce).
  • the plap enzymatic activity of the cell extracts is evaluated spectrophotometrically (alkaline phosphatase kit, BIORAD).
  • the plap units are determined in relation to a standard consisting of alkaline phosphatase from calf intestine (Boehringer Mannheim).
  • the neo activities are measured by the transfer of phosphate labeled with [ ⁇ - 32 P] on neomycin (Ramesh and Osborne, 1991, Anal. Biochem. 193, 316-318).
  • the results of the expression of the plap and neo genes measured in the absence and in the presence of rapamycin are illustrated in FIG. 4 and presented in Table 3.
  • Table 3 effect of rapamycin on the expression of plap and neo cistrons expressed in% relative to untreated cells.
  • rapamycin reduces the cap-dependent translation and increases that dependent on TIRES. This stimulation can be explained by less competition for the translational machinery in the presence of rapamycin.
  • the addition of rapamycin is accompanied by an increase in the expression of the two genes.
  • quantitative expression data indicate that the relative activity of IRES is different, suggesting a competition between them for ribosomes.
  • Dev cell line which constitutes a cellular model of the central nervous system, a potential target for human gene therapy.
  • Dev cells are derived from a human primary tumor of neuroectodermal origin (PNET) and behave like pluripotent stem cells (Derrington et al., 1997, Oncogene, in press). Moreover, it is possible to induce their differentiation either into neurons or into glial cells (Derrington et al., 1997, supra; Dufay et al., 1994, Eur. J. Neurosci. 6, 1633-1640; Giraudon et al ., 1993, Neurosci. 52, 1069-1079).
  • the dicistronic vector pREV HW-3 (IRES REV-A 265-578) is compared with the control vector pEMCV CBTV (IRES EMCV).
  • the Dev cells are infected with a 1/100 dilution of viral supernatant for 2 days then fixed, histochemically stained according to the above protocol and counted.
  • the viral titer is similar for the two vectors and of the order of 3 ⁇ 10 3 TU / ml.
  • the transduction units (TU / ml) correspond to the ratio of the number of colonies x dilution of the infecting retrovirus by the total volume (ml) of the diluted vector placed on the cells.
  • the target cells are as previously transduced with a 1/100 dilution of viral supernatant and selected in the presence of G418 for 3 weeks.
  • the intensity of the fluorescence is determined by flow cytometry with an antibody specific for the plap (DAKO). It is indicated that a polyclonal antibody is suitable.
  • the results show the production of the enzyme plap by cells transduced and selected with G418. In parallel, the synthesis of the enzyme plap is confirmed by histochemical staining of the resistant clones.
  • Variants are the cell culture conditions in the presence or absence of serum and the presence or absence of growth factors (FGF-2) and the production of viruses in the presence or absence of serum.
  • FGF-2 growth factors
  • Protocol 1 cells cultured in medium with serum (10%), virus produced in the presence of serum (10%).
  • Protocol 2 cells cultured in medium with serum (10%), virus produced in the absence of serum.
  • Protocol 3 cells cultured in serum-free medium, virus produced in the presence of serum (10%).
  • Protocol 4 cells cultured in serum-free medium, virus produced in the absence of serum.
  • Protocol 5 cells cultured in serum-free medium, virus produced in the presence of serum (10%), presence of FGF-2.
  • Protocol 6 cells cultured in serum-free medium, virus produced in the absence of serum, presence of FGF-2.
  • the percentage of Dev cells transduced is determined by flow cytometry ( Figure 5). The percentage of cells transduced by pREV HW-3 exceeds 30% when the culture of Dev cells is carried out in the absence of serum. Such a% is not reached with the conventional virus pEMCV CBTV. The addition of growth factors is also advantageous. Among all the protocols used, protocol 5 allows more than 50% of Dev cells to be transduced.
  • the expression of plap and neo cistrons is evaluated after neuronal and glial differentiation (Derrington et al., 1997, supra). Briefly, the Dev cells adopt a differentiated phenotype in the presence of serum and FGF-2 whereas when the culture is carried out in the absence of serum, the phenotype is pluripotent. Immunofluorescence results with a specific anti-plap antibody on transduced Dev cells, selected with G418 and differentiated show expression of the plap in neuronal and glial cells. These data suggest that the state of differentiation does not inhibit TIRES REV-A mediated translation.
  • TYPE OF MOLECULE RNA (genomics)
  • HYPOTHETIC NO
  • ANTI-SENSE NO
  • ORGANISM Reticuloendotheliosis virus
  • TYPE OF MOLECULE RNA (genomics)
  • HYPOTHETIC NO
  • ANTI-SENSE NO
  • ORGANISM Reticuloendotheliosis virus

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Communicable Diseases (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Plant Pathology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Vascular Medicine (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
EP98922883A 1997-04-28 1998-04-28 Neue interne ribosomenzugangsstelle und diese beinhaltender vektor Withdrawn EP0918875A1 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
FR9705203A FR2762615B1 (fr) 1997-04-28 1997-04-28 Nouveau site interne d'entree des ribosomes et vecteur le contenant
FR9705203 1997-04-28
PCT/FR1998/000849 WO1998049334A1 (fr) 1997-04-28 1998-04-28 Nouveau site interne d'entree des ribosomes et vecteur le contenant

Publications (1)

Publication Number Publication Date
EP0918875A1 true EP0918875A1 (de) 1999-06-02

Family

ID=9506363

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98922883A Withdrawn EP0918875A1 (de) 1997-04-28 1998-04-28 Neue interne ribosomenzugangsstelle und diese beinhaltender vektor

Country Status (7)

Country Link
US (1) US6783977B1 (de)
EP (1) EP0918875A1 (de)
JP (1) JP2001500021A (de)
AU (1) AU749250B2 (de)
CA (1) CA2258490A1 (de)
FR (1) FR2762615B1 (de)
WO (1) WO1998049334A1 (de)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2766091A1 (fr) 1997-07-18 1999-01-22 Transgene Sa Composition antitumorale a base de polypeptide immunogene de localisation cellulaire modifiee
EP1266022B1 (de) * 2000-03-24 2008-10-22 Cell Genesys, Inc. Interne ribosomenzugangsstelle-enthaltende zell-spezifische adenovirale vektoren
PT1649023E (pt) 2003-07-21 2008-11-20 Transgene Sa Polipéptido com actividade de citosina desaminase melhorada
US20080096228A1 (en) * 2006-08-08 2008-04-24 The Regents Of The University Of California Compositions And Methods Relating To Mammalian Internal Ribosome Entry Sites
ATE498008T1 (de) 2006-10-23 2011-02-15 Res Org Information & Systems Verfahren zur herstellung eines zellstamms mit fähigkeit zur tetracyclin-induzierten genexpression oder eines zellstamms mit konditionalem gen-knockout sowie verwendung der zellstämme
WO2010054379A2 (en) 2008-11-10 2010-05-14 The United States Of America, As Represensted By The Secretary, Department Of Health And Human Services Gene signature for predicting prognosis of patients with solid tumors
US9150926B2 (en) 2010-12-06 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Diagnosis and treatment of adrenocortical tumors using human microRNA-483
CA2835825C (en) 2011-05-13 2021-03-23 Minoru S.H. Ko Use of zscan4 and zscan4-dependent genes for direct reprogramming of somatic cells
CA3176712A1 (en) 2013-03-15 2014-09-18 Elixirgen Therapeutics, Inc. Methods of using zscan4 for rejunevating human cells
CN114807145B (zh) * 2022-06-02 2024-02-20 新乡医学院 一种提高非帽子依赖翻译效率的前导序列及其应用

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5112767A (en) * 1988-03-04 1992-05-12 University Of Southern California Vectors with enhancer domains
US5997856A (en) * 1988-10-05 1999-12-07 Chiron Corporation Method and compositions for solubilization and stabilization of polypeptides, especially proteins
WO1993003143A1 (en) * 1991-08-07 1993-02-18 Anderson W French Retroviral vectors containing internal ribosome entry sites
GB9119762D0 (en) * 1991-09-16 1991-10-30 Filler Aaron G Particulate agents for nmt
DE4228458A1 (de) * 1992-08-27 1994-06-01 Beiersdorf Ag Multicistronische Expressionseinheiten und deren Verwendung
DE4228457A1 (de) * 1992-08-27 1994-04-28 Beiersdorf Ag Herstellung von heterodimerem PDGF-AB mit Hilfe eines bicistronischen Vektorsystems in Säugerzellen
WO1994029437A1 (en) * 1993-06-07 1994-12-22 University Of Medicine & Dentistry Of New Jersey Highly-efficient, self-inactivating, recombination-free, u3-free retroviral vectors
FR2722208B1 (fr) * 1994-07-05 1996-10-04 Inst Nat Sante Rech Med Nouveau site interne d'entree des ribosomes, vecteur le contenant et utilisation therapeutique
US5665350A (en) * 1994-11-23 1997-09-09 University Of Massachusetts Medical Center Cell cycle dependent transplantation and ex vivo gene therapy
US6136566A (en) * 1996-10-04 2000-10-24 Lexicon Graphics Incorporated Indexed library of cells containing genomic modifications and methods of making and utilizing the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9849334A1 *

Also Published As

Publication number Publication date
JP2001500021A (ja) 2001-01-09
CA2258490A1 (fr) 1998-11-05
FR2762615A1 (fr) 1998-10-30
US6783977B1 (en) 2004-08-31
FR2762615B1 (fr) 1999-07-16
AU7536598A (en) 1998-11-24
WO1998049334A1 (fr) 1998-11-05
AU749250B2 (en) 2002-06-20

Similar Documents

Publication Publication Date Title
EP0784693B1 (de) Virale vektoren für die gentherapie
FR2722208A1 (fr) Nouveau site interne d'entree des ribosomes, vecteur le contenant et utilisation therapeutique
EP0842279B1 (de) Hilfsvirus für die herstellung von rekombinanten virusvektoren
EP0988391B1 (de) Rekombinante adenovirale vektoren, die eine spleissequenz enthalten
WO1998002529A1 (fr) Lignees d'encapsidation hautement productrices
EP0918875A1 (de) Neue interne ribosomenzugangsstelle und diese beinhaltender vektor
EP0781344B1 (de) Implantat und vektor zur behandlung von erworbenen krankheiten
EP1078094B1 (de) Methoden und zusammensetzungen für die produktion von viruspartikeln
EP1002120B1 (de) Chimäre adenovirale vektoren
EP1108051A2 (de) Induzierbares expresionssystem
EP0906443B1 (de) In vivo herstellung von replicativen molekülen
EP0796338A1 (de) Verpakungszellinie zur transkomplementierung von defektiven retroviren-vektoren
WO1997044475A1 (fr) Vecteurs adenoviraux pour la therapie genique
FR2737221A1 (fr) Nouveaux vecteurs viraux pour la therapie genique
Vile et al. Retroviral Vectors From Laboratory Tools to Molecular Medicines
Yu et al. High Efficiency Retroviral Vectors with Improved Safety
WO1998049335A1 (fr) Construction et expression d'enveloppe chimerique de retrovirus par des vecteurs, et compositions pharmaceutiques les contenant

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

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 MC NL PT SE

17P Request for examination filed

Effective date: 19990426

17Q First examination report despatched

Effective date: 20070328

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

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20090302