WO2000018908A1 - Utilisation de promoteurs specifiques hybrides pour controler l'expression tissulaire - Google Patents

Utilisation de promoteurs specifiques hybrides pour controler l'expression tissulaire Download PDF

Info

Publication number
WO2000018908A1
WO2000018908A1 PCT/FR1999/002265 FR9902265W WO0018908A1 WO 2000018908 A1 WO2000018908 A1 WO 2000018908A1 FR 9902265 W FR9902265 W FR 9902265W WO 0018908 A1 WO0018908 A1 WO 0018908A1
Authority
WO
WIPO (PCT)
Prior art keywords
promoter
gene
smooth muscle
region
cells
Prior art date
Application number
PCT/FR1999/002265
Other languages
English (en)
French (fr)
Inventor
Didier Branellec
Raphaël Darteil
Abderrahim Mahfoudi
Daniel Scherman
Original Assignee
Aventis Pharma S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from FR9812000A external-priority patent/FR2783839B1/fr
Application filed by Aventis Pharma S.A. filed Critical Aventis Pharma S.A.
Priority to CA002343922A priority Critical patent/CA2343922A1/fr
Priority to HU0105230A priority patent/HUP0105230A3/hu
Priority to AU56324/99A priority patent/AU775717B2/en
Priority to IL14210799A priority patent/IL142107A0/xx
Priority to JP2000572355A priority patent/JP2002525109A/ja
Priority to EP99943036A priority patent/EP1115857A1/fr
Priority to KR1020017003768A priority patent/KR20010075338A/ko
Publication of WO2000018908A1 publication Critical patent/WO2000018908A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4716Muscle proteins, e.g. myosin, actin
    • 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
    • C12N2710/10345Special targeting system for viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination

Definitions

  • the present invention relates to the field of biology, and in particular to the field of regulation of gene expression. It describes in particular new constructions and new vectors allowing a targeted and strong expression of genes.
  • the present invention can be used in many fields, and in particular for the production of recombinant proteins, for the creation of transgenic animal models, for the creation of cell lines, for the development of screening tests, or even in gene therapy. and cellular.
  • the ability to control and direct gene expression is a very important issue in the development of biotechnology. In vitro, it can make it possible to improve the conditions for the production of recombinant proteins, by using specific populations of cells. Still in vitro, it can allow the detection or the demonstration of the presence of specific populations of cells in a sample, or also to test the properties of a product or the regulation of a gene in a specific population of cells.
  • the control of gene expression is also very important for ex vivo or in vivo therapeutic approaches, in which the possibility of selectively controlling the production of a therapeutic molecule is essential. In fact, depending on the applications, depending on the gene to be transferred, it is important to be able to target certain tissues or only certain parts of an organism in order to concentrate the therapeutic effect and limit dissemination and side effects.
  • a first approach consists, for example, in using vectors or transfer agents having a specific cell specificity.
  • the specificity conferred by this type of vector is generally quite coarse and does not allow targeting of precise populations of cells.
  • Another approach is to use expression signals specific for certain cell types.
  • so-called "specific" promoters have been described in the literature, such as the promoter of genes coding for pyruvate kinase, villin, GFAP, the promoter of the intestinal fatty acid binding protein, the ⁇ -actin promoter of smooth muscle cells, the SM22 promoter or also the promoter of the human albumin gene for example.
  • promoters have tissue specificity, they also have, in return, a relatively low potency.
  • tissue specificity the vast majority of these promoters have activity levels which are well below those of so-called "strong" promoters, generally by a factor of between 10 and 100 at least.
  • specificity of a promoter is inversely proportional to its strength and that, the more the strength is increased, the higher the level of non-specific activity.
  • the object of the present invention is precisely to provide new constructions allowing the strong and targeted expression of genes.
  • the invention describes in particular new chimeric promoters allowing a strong and specific gene expression of smooth muscle cells.
  • the invention also describes vectors containing such promoters and their use for the transfer of genes into cells in vitro, ex vivo and in vivo.
  • the constructs of the invention make it possible for the first time to combine opposite properties, namely high selectivity and high transcriptional activity.
  • the present invention thus provides a particularly efficient means for targeting the expression of genes in smooth muscle cells, in vivo or in vitro, and for regulating this expression.
  • the invention is based more particularly on the construction of chimeric (or hybrid) promoters, comprising regions of origin and of different function. More particularly, a first object of the invention resides in a hybrid promoter comprising:
  • enhancer regions and promoters have already been described in the prior art.
  • non-specific promoters such as the promoter of the chicken ⁇ -actin gene (WO96 / 13597) in order to increase their strength.
  • WO96 / 13597 non-specific promoters
  • Such constructions have not been described or suggested for the purpose of attempting to obtain a strong and specific expression of smooth muscle cells.
  • the invention is based in part on the selection and combination of specific "enhancer” elements and specific "promoter” elements.
  • the invention is also based on the demonstration that this combination of elements makes it possible to obtain expression at high levels, without affecting the selectivity of the promoter for the target cells of smooth muscle.
  • the invention therefore provides particularly advantageous constructions since they allow targeted production and with high levels of molecules in smooth muscle cells.
  • the present application also shows that these constructs can be used both in vitro and in vivo.
  • the enhancer region and the promoter region are associated in a functional manner, that is to say so that the enhancer region exerts a stimulating activity on the activity of the promoter region.
  • these two regions are therefore genetically linked and are close enough to each other to allow the enhancer region to activate the promoter region.
  • the distance separating the enhancer region and the promoter region is less than 1 kb, more preferably less than 500 bp.
  • these two regions are spaced less than 400 bp, more preferably less than 200 bp.
  • the respective orientation of the two regions has no significant influence on the activity of the hybrid promoters of the invention.
  • the enhancer region can be positioned in the same orientation or in the reverse orientation with respect to the direction of transcription of the promoter region.
  • the enhancer region is chosen from the enhancer region of the immediate early cytomegalovirus gene (CMV-IE), the enhancer region of the red sarcoma virus LTR (LTR-RSV), the enhancer region SV40 virus, and the enhancer region of the EF1 ⁇ gene. More preferably, in the hybrid promoters of the invention, the enhancer region is the enhancer region of the immediate early gene of the cytomegalovirus (CMV-IE), preferably of the human cytomegalovirus (hCMV-IE).
  • a particular enhancer region consists for example of the fragment -522 to -63 of the hCMV-IE gene, or of any fragment comprising at least part of it and exhibiting enhancer activity.
  • a region comprising all or part of the promoter of the gene coding for ⁇ -actin of smooth muscle cells (SMact) or of the SM22 gene is advantageously used for implementing the invention.
  • the promoter of these genes has been described for its specific character of smooth muscle cells (see in particular Ueyama H. et al., Mol. Cell. Biol, 4 (1984) 1073-1078; Solway J. et al., J. Biol. Chem., 270 (1995) 13460-13469).
  • SMact smooth muscle cells
  • the promoter region used is a chimeric region comprising a basal promoter and a sequence conferring tissue specificity, said sequence being derived from the SMact promoter or from the SM22 promoter, or from a combination of the two.
  • the basal promoter can be a "minimal" promoter, that is to say comprising only the sequences essential to the activity of transcriptional promoter (for example a TATA box).
  • This basal promoter can be the own basal promoter of the SMact or SM22 gene, or of heterologous origin ( ⁇ -globin, HSV-TK, SV40 or EF1- ⁇ for example).
  • the sequence conferring tissue specificity advantageously comprises part of the sequence of the SMact promoter (RT Shimizu et al. J. Biol. Chem. 270 (1995) 7634-7643) and / or of the SM22 promoter (L. Li et al. J Cell Biol. 132 (1996) 849-859; S. Kim et al. J. Clin. Invest. 100 (1997) 1006-1014; Kemp et al. Biochem. J. 310 (1995) 1037-1043).
  • a particular type of hybrid promoter according to the invention therefore comprises:
  • a sequence conferring tissue specificity comprising all or part of the SMact promoter and / or the SM22 promoter.
  • the enhancer region and the promoter region can be isolated by conventional techniques from nucleic acid libraries or from total cellular DNA, for example by amplification by means of specific probes. These fragments can also be synthesized artificially using the sequence information available in the prior art. When these fragments are obtained, they can be easily combined with one another by means of ligases and other restriction enzymes, to generate hybrid promoters of the invention. In addition, these fragments can be modified by digestion, mutation, insertion or addition of base pairs, either in order to facilitate their cloning, or in order to modify their functional properties.
  • the fragments can be associated directly with one another or, on the contrary, spaced by base pairs having no significant influence on the activity of the hybrid promoter.
  • the hybrid promoters of the invention thus have the capacity to express a nucleic acid of interest in a specific manner in smooth muscle cells.
  • the "specific" character of the expression means that the promoter activity is significantly very higher in the smooth muscle cells. Although a non-specific expression can exist in other cells, the corresponding level of activity generally remains very low (negligible) compared to that observed in smooth muscle cells, generally at least a factor of 10 .
  • the results presented in the examples show in this respect an expression differential which can reach a factor of 140, which testifies to the high selectivity of the promoters of the invention.
  • results presented also show a high specificity with regard to smooth muscle cells since no expression was detected in the endothelial cells which are located in the vicinity, in the blood vessel, in particular the artery.
  • the results presented in the examples also show that the strength of the promoters of the invention is much greater than that of specific non-hybrid promoters, the differential being able to exceed a factor of 100.
  • another subject of the invention relates to an expression cassette comprising a nucleic acid coding for an RNA or a polypeptide of interest, placed under the control of a hybrid promoter as defined above.
  • the cassette of the invention further comprises a transcription termination signal, placed 3 'to the nucleic acid.
  • the nucleic acid can code, for example, for a protein chosen from: proteins involved in the cell cycle, such as for example p21 or any other kinase inhibitor protein cyclin-dependent (cdk), the retinoblastoma (Rb) gene product, GAX, GAS-1, GAS-3, GAS-6, Gadd 45, Gadd 153, cyclins A, B and D.
  • apoptosis such as for example p53, members of the family of apoptosis inducers such as Bas, Bcl-X s , Bad or any other antagonist of Bcl 2 and of Bcl-Xi.
  • proteins capable of modifying the proliferation of smooth muscle cells such as for example an intracellular antibody or an ScFv inhibiting the activity of proteins involved in cell proliferation, such as for example the Ras protein, mapkinase, or receptors for tyrosine kinase or growth factors.
  • proteins inducing angiogenesis such as for example members of the VEGF family, members of the FGF family and more particularly FGF1, FGF2, FGF4, FGF5, angiogenin, EGF, TGF ⁇ , TGF ⁇ , TNF ⁇ , Scatter Factor / HGF , members of the angiopoetins family, cytokines and in particular interleukins including IL-1, IL-2, IL-8, angiotensin-2, plasminogen activator (TPA), urokinase (uPA) , the molecules involved in the synthesis of active lipids (prostaglandins, Cox-1).
  • proteins inducing angiogenesis such as for example members of the VEGF family, members of the FGF family and more particularly FGF1, FGF2, FGF4, FGF5, angiogenin, EGF, TGF ⁇ , TGF ⁇ , TNF ⁇ , Scatter Factor / HGF , members of the angiopoetins family, cytokines and in particular inter
  • transcription factors such as for example natural or chimeric nuclear receptors, comprising a DNA binding domain, a ligand binding domain and a transcription activating or inhibiting domain, such as for example fusion proteins tetR-NLS-VP16, estrogen receptor fusion proteins, steroid hormone receptor fusion proteins, progesterone receptor fusion proteins, CID (Chemical Inducer of) proteins Dimerization) described by Rivera et al. (Rivera et al. (1996), A humanized
  • the present invention is not limited to particular examples of proteins or RNA, but that it can be used by a person skilled in the art for the expression of any nucleic acid in smooth muscle cells, for example. simple usual experimental operations.
  • the vector of the invention can be for example a plasmid, a cosmid or any DNA not encapsulated by a virus, a phage, an artificial chromosome, a recombinant virus, etc. It is preferably a plasmid or a recombinant virus.
  • plasmid type vectors mention may be made of all the cloning and / or expression plasmids known to those skilled in the art and which generally have an origin of replication. Mention may also be made of new generation plasmids carrying origins of replication and / or improved markers as described for example in applications WO96 / 26270 and PCT / FR96 / 01414.
  • adenovirus viruses there may preferably be mentioned adenovirus viruses, retroviruses, herpes virus or recombinant adeno-associated viruses.
  • the construction of this type of recombinant virus defective for replication has been widely described in the literature, as well as the infection properties of these vectors (see in particular S. Baeck and KL March (1998), Circul. Research vol. 82, pp 295-305), T. Shenk, BN Fields, DM Knipe, PM Howley et al (1996), Adenoviridae: the viruses and their replication (in virology).
  • Pp 211-2148 EDS - Ravenspublishers / Philadelphia, P. Yeh and M. Perricaudet (1997), FASEB Vol. 11, pp 615-623.
  • a particularly preferred recombinant virus for the implementation of the invention is a defective recombinant adenovirus.
  • Adenoviruses are linear double-stranded DNA viruses around 36 (kilobases) kb in size. There are different serotypes, including the structure and properties vary somewhat, but have a comparable genetic organization. More particularly, the recombinant adenoviruses can be of human or animal origin. As regards adenoviruses of human origin, mention may preferably be made of those classified in group C, in particular adenoviruses of type 2 (Ad2), 5 (Ad5), 7 (Ad7) or 12 (Adl2).
  • adenoviruses of animal origin mention may preferably be made of adenoviruses of canine origin, and in particular all the strains of the adenovirus CAV2 [Manhattan strain or A26 / 61 (ATCC VR-800) for example].
  • Other adenoviruses of animal origin are cited in particular in application WO94 / 26914 incorporated herein by reference.
  • the adenovirus genome includes in particular a repeated inverted sequence (ITR) at each end, an encapsidation sequence (Psi), early genes and late genes.
  • ITR inverted sequence
  • Psi encapsidation sequence
  • the main early genes are contained in the E1, E2, E3 and E4 regions. Among these, the genes contained in the El region in particular are necessary for viral propagation.
  • the main late genes are contained in regions L1 to L5.
  • the genome of the Ad5 adenovirus has been fully sequenced and is accessible on the database (see in particular Genebank M73260). Likewise, parts or even all of other adenoviral genomes (Ad2, Ad7, Ad 12, etc.) have also been sequenced.
  • adenovirus For their use as recombinant vectors, various constructs derived from adenoviruses have been prepared, incorporating different therapeutic genes. In each of these constructions, the adenovirus was modified so as to render it incapable of replication in the infected cell. Thus, the constructions described in the prior art are deleted adenoviruses from the E1 region, essential for viral replication, at the level of which heterologous DNA sequences are inserted (Levrero et al, Gene 101 (1991) 195; Gosh -Choudhury et al., Gene 50 (1986) 161). Furthermore, to improve the properties of the vector, it has been proposed to create other deletions or modifications in the genome of the adenovirus.
  • thermosensitive point mutation was introduced into the mutant ts125, making it possible to inactivate the 72kDa DNA binding protein (DBP) (Van der Vliet et al., 1975).
  • Other vectors include a deletion from another region essential for replication and / or viral propagation, the E4 region.
  • the E4 region is in fact involved in the regulation of the expression of late genes, in the stability of late nuclear RNA, in the extinction of the expression of proteins of the host cell and in the efficiency of replication of l 'Viral DNA.
  • Adenoviral vectors in which the E1 and E4 regions are deleted therefore have very reduced transcription background noise and expression of viral genes.
  • Such vectors have been described by example in applications WO94 / 28152, WO95 / 02697, WO96 / 22378).
  • vectors carrying a modification in the IVa2 gene have also been described (WO96 / 10088).
  • the recombinant adenovirus is a human adenovirus of group C. More preferably, it is an adenovirus Ad2 or Ad5.
  • the recombinant adenovirus used in the context of the invention comprises a deletion in the E1 region of its genome. Even more particularly, it includes a deletion of the Ela and Elb regions. As a specific example, mention may be made of deletions affecting nucleotides 454-3328; 382-3446 or 357-4020 (with reference to the Ad5 genome).
  • the recombinant adenovirus used in the context of the invention further comprises a deletion in the E4 region of its genome. More particularly, the deletion in the E4 region affects all of the open phases. As a specific example, the deletions 33466-35535 or 33093-35535 can be cited. Other types of deletions in the E4 region are described in applications WO95 / 02697 and WO96 / 22378, incorporated herein by reference.
  • the expression cassette can be inserted at different sites of the recombinant genome. It can be inserted at the level of the E1, E3 or E4 region, replacing the deleted or surplus sequences. It can also be inserted at any other site, apart from the sequences necessary in cis for the production of viruses (ITR sequences and packaging sequence).
  • Recombinant adenoviruses are produced in an packaging line, that is to say a cell line capable of complementing in trans one or more of the deficient functions in the recombinant adenoviral genome.
  • packaging lines known to those skilled in the art, mention may be made, for example, of line 293 in which a part of the adenovirus genome has been integrated.
  • line 293 is a human embryonic kidney cell line containing the left end (approximately 11-12%) of the genome of the adenovirus serotype 5 (Ad5), comprising the left ITR, the packaging region , the El region, including Ela and Elb, the region coding for the pIX protein and part of the region coding for the pIVa2 protein.
  • This line is capable of trans-complementing recombinant adenoviruses defective for the E 1 region, that is to say devoid of all or part of the E1 region, and of producing viral stocks having high titers.
  • This line is also capable of producing, at permissive temperature (32 ° C.), stocks of virus further comprising the thermosensitive E2 mutation.
  • Recombinant adenoviruses are usually produced by the introduction of viral DNA into the packaging line, followed by lysis of the cells after approximately 2 or 3 days (the kinetics of the adenoviral cycle being 24 to 36 hours).
  • the viral DNA introduced may be the complete recombinant viral genome, optionally constructed in a bacterium (WO96 / 25506) or in a yeast (WO95 / 03400), transfected in the cells. It can also be a recombinant virus used to infect the packaging line.
  • the viral DNA can also be introduced in the form of fragments each carrying a part of the recombinant viral genome and a zone of homology allowing, after introduction in the packaging cell, to reconstitute the recombinant viral genome by homologous recombination between the different fragments.
  • the recombinant viral particles are isolated by centrifugation in a cesium chloride gradient.
  • An alternative method has been described in application FR96: 08164 incorporated herein by reference.
  • the invention also relates to a composition
  • a composition comprising a vector as defined above and a chemical or biochemical transfer agent.
  • chemical or biochemical transfer agent is understood to mean any compound (i.e., other than a recombinant virus) facilitating the penetration of a nucleic acid into a cell.
  • They can be cationic non-viral agents such as cationic lipids, peptides, polymers (Polyethylene Imine, Polylysine), nanoparticles; or non-cationic non-viral agents such as non-cationic liposomes, polymers or non-cationic nanoparticles.
  • cationic non-viral agents such as cationic lipids, peptides, polymers (Polyethylene Imine, Polylysine), nanoparticles
  • non-cationic non-viral agents such as non-cationic liposomes, polymers or non-cationic nanoparticles.
  • Such agents are well known to those skilled in the art.
  • the invention also relates to a composition
  • a composition comprising a recombinant virus as defined above and a physiologically acceptable vehicle.
  • the invention also relates to a pharmaceutical composition comprising a vector as described above.
  • the pharmaceutical 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, in particular for an intravascular injection or in the tissues of smooth muscle.
  • 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, through addition according to the case of sterilized water or physiological saline, allow the constitution of injectable solutes.
  • Other excipients can be used such as for example a hydrogel.
  • This hydrogel can be prepared from any biocompatible and non-cytotoxic polymer (homo or hetero). Such polymers have for example been described in application WO93 / 08845.
  • the use of a hydrogel is particularly advantageous for the transfer of nucleic acids into the vascular walls, and in particular into the smooth muscle cells of the vascular walls.
  • the doses used for the injection can be adapted according to different parameters, and in particular according to the mode of administration used, the aim pursued (labeling, pathology, screening, etc.), the gene to be expressed, or even the duration of the expression sought.
  • the recombinant viruses according to the invention are formulated and administered in the form of doses of between 10 ⁇ and 10 ⁇ pfu, and preferably 10 ⁇ to 10 ⁇ pfu.
  • pfu plaque forming unit
  • plaque forming unit corresponds to the infectious power of a viral solution, and is determined by infection of an appropriate cell culture, and measurement of 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 cassettes, vectors or compositions of the invention can be incubated at conventional doses in the presence of the selected cell populations. These incubations can be carried out on culture dishes, flasks, fermenters, or any other device chosen.
  • the invention also relates to any cell modified by a cassette or a vector (in particular an adenovirus) as described above.
  • modified cell is understood to mean any cell containing a construct according to the invention. These cells can be used for the production of recombinant proteins in vitro. They can also be intended for implantation in an organism, according to the methodology described in application WO95 / 14785. These cells are preferably human smooth muscle cells.
  • the invention also relates to the use of a hybrid promoter as defined above for the specific expression of a nucleic acid in smooth muscle cells, in vitro, ex vivo or in vivo.
  • the invention also relates to the use of a hybrid promoter as defined above for the preparation of a composition intended for the expression of a nucleic acid in smooth muscle cells in vivo and not in endothelial cells which are in the vicinity in the artery.
  • the constructs according to the invention can also be used for the creation of animal models of vascular pathologies or for carrying out labeling studies or in methods of detecting or screening for presence of smooth muscle cells in samples.
  • the present invention also relates to a method for producing recombinant proteins comprising the introduction into a cell population of a vector as defined above, the culture of said recombinant cell population, and the recovery of said protein produced.
  • smooth muscle cells are used for the implementation of the method of the invention. These can be established lines or primary cultures.
  • Table I Relative activities of hybrid promoters (hSM ⁇ -actin) evaluated in transient transfections in vitro in rabbit smooth muscle cells in primary culture (rabbit SMC), in ECV304 cells, in C2C12 myoblasts, in HeLa cells, in NIH 3T3 cells, in TU182 carcinoma cells, as well as in renal 293 cells.
  • the relative activity of each promoter is expressed as a percentage of luciferase activity obtained with the plasmid pCMV-leadTK.
  • Enh-X the enhancer sequence of hCMV-IE is cloned upstream of promoter X according to its normal orientation.
  • HnE-X the enhancer sequence of hCMV-IE is cloned upstream of promoter X in the opposite orientation.
  • Table II Relative activities of hybrid promoters (mSM22) evaluated in transient transfections in vitro in rabbit smooth muscle cells in primary culture (rabbit SMC), in ECV304 cells, in C2C12 myoblasts, in HeLa cells, in cells NIH 3T3, in TU182 carcinoma cells, as well as in renal cells 293.
  • the relative activity of each promoter is expressed as a percentage of the luciferase activity obtained with the plasmid pCMV-leadTK.
  • Enh-X the enhancer sequence of hCMV-IE is cloned upstream of promoter X according to its normal orientation.
  • HnE-X the enhancer sequence of hCMV-IE is cloned upstream of promoter X in the opposite orientation.
  • Figure 1 Schematic representations of the plasmids whose expression cassette contains the hybrid promoter hSM ⁇ -actin.
  • Figure 2 Schematic representations of the plasmids whose expression cassette contains the hybrid promoter mSM22 ⁇ .
  • Figure 3 Activities of hybrid promoters evaluated in transient transfections in vitro in rabbit smooth muscle cells in primary culture (rabbit SMC), in endothelial cells derived from human umbilical cord carcinoma (ECV304), in myoblasts of mice (C2C12) as well as in epithelial cells from a carcinoma of the human cervix (HeLa).
  • the relative activity of each promoter is expressed as a percentage of the luciferase activity obtained with the plasmid pCMV-leadTK.
  • Enh-X the enhancer sequence of hCMV-IE is cloned upstream of promoter X according to its normal orientation.
  • hnE-X the enhancer sequence of hCMV-IE is cloned upstream of promoter X in the opposite orientation.
  • Figure 4 Activities of hybrid promoters evaluated in transient transfections in vitro in mouse embryonic fibroblasts (NIH 3T3), in cells derived from a human ENT carcinoma (TU182), as well as in transformed human embryonic renal cells (293) . The relative activity of each promoter is expressed as a percentage of the luciferase activity obtained with the plasmid pCMV-leadTK.
  • Enh-X the enhancer sequence of hCMV-IE is cloned upstream of promoter X according to its normal orientation.
  • hnE-X the enhancer sequence of hCMV-IE is cloned upstream of promoter X in the opposite orientation.
  • Figure 5 Activities of hybrid promoters evaluated in gene transfer in vivo in the cranial tibial muscle of C57BL6 mice. The relative activity of each promoter is expressed as a percentage of the luciferase activity obtained with the plasmid pCMV-leadTK.
  • the plasmid pGL3 -Basic used for cloning the different promoter regions, is of commercial origin (Promega Corporation).
  • the plasmids pCMV ⁇ (Clontech Laboratories Inc.) and pUC18 (Boehringer Mannheim) are also of commercial origin.
  • the enzymatic amplification of DNA fragments by the PCR technique can be carried out using a DNA thermal cycler ⁇ (Perkin Elmer Cetus) according to the manufacturer's recommendations.
  • the electroporation of plasmid DNA in Escherichia coli cells can be carried out using an electroporator (Bio-Rad) according to the manufacturer's recommendations.
  • 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 Applied Biosystems according to the manufacturer's recommendations.
  • EXAMPLE 1 Construction of hybrid promoters and expression plasmids containing them.
  • PhSMact plasmid The high molecular weight genomic DNA was prepared according to the method described by Sambrook et al. ("Molecular Cloning, a Laboratory Manual", Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989) from a primary culture of human aortic smooth muscle cells (Clonetics).
  • This DNA was used as a template for a first amplification by PCR using the following primers:
  • - Primer 6417 (5 'GATGGTCCCTACTTATGCTGCTA 3') (SEQ ID 1) starting at position -1034 (promoter region) of the human specific smooth muscle ⁇ -actin gene (Ueyama H. et al., Mol. Cell. Biol, 4 (1984) 1073-1078. Access Genbank D00618).
  • - Primer 6418 (5 'CTTCCATCATACCAAACTACATA 3') (SEQ ID 2) at position 1974 of the sequence D00618 located inside the first intron of the hSMact gene.
  • the reaction mixture comprises 1 mg of genomic DNA, 10 pmol of each of the two primers (6417 and 6418), 100 mM of each deoxyribonucleotide (dATP, dCTP, dGTP, dTTP), 2 mM MgC12 and 5 units of Taq DNA polymerase ( PerkinElmer).
  • the reaction volume is made up to 50 ml adjusted to the optimal concentration of the ACP buffer recommended by Perkin Elmer.
  • the PCR amplification is carried out in Micoamp TM tubes (Perkin Elmer) using a PTC-100 TM thermocycler (MJ Research, Inc.).
  • This amplification consists of a denaturation step at 95 ° C for 2 min followed by 30 cycles including a denaturation step of 15 sec at 95 ° C, a hybridization step of 30 sec at 60 ° C and an extension step 1 min at 72 ° C. These thirty cycles are followed by an additional extension of 5 min then the ACP reactions are stored at 10 ° C.
  • Primer 6453 (5 'CTGCTAAATTGctcgagGACAAATTAGACAAA 3 * ) (SEQ ID 3), this primer introduces an Xhol site (lowercase underlined) upstream of the hSMact promoter (position -680).
  • the DNA fragment amplified by PCR is digested for 3 hours at 37 ° C. with Xhol and HindIII and then cloned into the vector pGL3-Basic (Promega) previously digested with these same enzymes. restriction, to generate the plasmid phSMact ( Figure 1).
  • Plasmids pXL3130 and pXL3131 A DNA fragment corresponding to the enhancer region of the promoter of the human cytomegalovirus IE gene (hCMV-IE) between the positions -522 and -63 relative to the site of initiation of the transcription, was amplified by PCR using the plasmid pCMV ⁇ as template and the oligonucleotides 8557 (5 'ATC GAC GCG TGC CCG TTA CAT AAC TTA CGG 3') (SEQ ID 5) and 8558 (5 'ATC GAC GCG TCC GCT CGA GCG TCA ATG GGG CGG AGT TG 3 ') (SEQ ID 6) as primers.
  • hCMV-IE human cytomegalovirus IE
  • This fragment was digested with MluI and then was cloned into the plasmid phSMact previously digested with MluI and treated with alkaline phosphatase.
  • two different plasmids were obtained: pXL3130 and pXL3131.
  • the schematic representations of these plasmids are gathered in the figure ( Figure 1).
  • These plasmids comprise, in the form of an Mlul-NDSl fragment, a hybrid promoter consisting of the enhancer of the promoter of the hCMV-IE gene and of the promoter of the hSM ⁇ -actin gene.
  • This DNA was used as a template for PCR amplification using the following primers:
  • this primer introduces an Xhol site (lowercase underlined) in position -436 of the promoter of the mouse SM22 alpha gene (Solway J. et al., J. Biol. Chem., 270 (1995) 13460-13469; Access Genbank L41161).
  • this primer introduces a HindIII site at position +43 of mSM22 alpha.
  • a reaction mixture comprising 1 mg of mouse genomic DNA, and 10 pmol of each of the two primers (6517 and 6518) was prepared with the same reagents as for hSMact and at the same concentrations, followed by PCR amplification carried out under the same conditions (see example 1.1.).
  • Plasmid pXL3152 and pXL3153 A DNA fragment corresponding to the enhancer region of the promoter of the hCMV-IE gene between the positions -522 and -63 relative to the transcription initiation site, was amplified by PCR using the plasmid pCMV ⁇ as template and oligonucleotides 8557 (5 'ATC GAC GCG TGC CCG TTA CAT AAC TTA CGG 3') and 8558 (5 'ATC GAC GCG TCC GCT CGA GCG TCA ATG GGG CGG AGT TG 3') (SEQ ID 6) as primers.
  • This fragment was digested with MluI and then was cloned into the plasmid pmSM22 previously digested with MluI and treated with alkaline phosphatase.
  • two different plasmids were obtained: pXL3152 and pXL3153.
  • the schematic representations of these plasmids are collated in FIG. 2.
  • These plasmids comprise, in the form of an Mlul-NDTl fragment, a hybrid promoter consisting of the enhancer of the promoter of the hCMV-IE gene and of the promoter of the mSM22 gene.
  • the expression vector pCGN previously described by Tanaka et al. contains the CMV promoter (-522 / + 72) fused to the "leader" of the HSV tk gene (+ 51 / + 101) upstream of a sequence coding for the epitope hemagglutinin.
  • Plasmid pCGN (10 ng) was used as a template for PCR amplification. The PCR reaction and the amplification were carried out under the same conditions as those used for hSMact and mSM22 (Examples 1.1 and 1.2).
  • primers that have been used are as follows: - Primer 6718 (5 'CCCGTTACATAACTTACGGTAAATGGCCCG 3') (SEQ ID 9), this primer hybridizes with the CMV promoter in position -522 (8 nucleotides downstream of the EcoRI site of pCGN).
  • the PCR fragment thus obtained is purified and then phosphorylated using the polynucleotide kinase from phage T4 (New England Biolabs).
  • the vector pGL3-Basic Promega was linearized with purified NcoL then treated with Klenow DNA polymerase (Boehringer Manheim) in order to fill the Ncol site.
  • Klenow DNA polymerase Boehringer Manheim
  • This vector is then dephosphorylated using alkaline phosphatase (Boehringer Manheim) and then used for the insertion of the phosphorylated PCR fragment.
  • the guanosine (G) of primer 6719 makes it possible to restore the Ncol site only when the CMV-leader tk fragment is oriented with the 5 ′ part (primer 6718, position -522 of CMV) downstream of the HindIII site of pGL3 -Basic and its 3 ′ end (primer 6719, leader tk) is ligated to the Ncol site of pGL3-Basic (first luciferase ATG).
  • the plasmid thus obtained is designated pCMV-leadTK.
  • This example illustrates the tissue specificity properties of the hybrid promoters of the invention in vitro.
  • SMC Rabbit smooth muscle cells
  • DMEM TM medium (Life Technologies Inc.) supplemented with 20%) of fetal calf serum (SVF).
  • ECV304 cells are cultured in 199 TM medium (Life Technologies Inc.) supplemented with 10%> of FCS.
  • C2C12 myoblasts, HeLa cells, NIH 3T3 cells and TU 182 cells are cultured in DMEM TM medium supplemented with 10% of SVF.
  • the 293 cells are cultured in MEM TM medium (Life Technologies Inc.) supplemented with pyruvate, non-essential amino acids and 10% FCS. All the cultures are carried out in an oven at 37 ° C., in a humid atmosphere and under a partial pressure of CO 2 of 5%.
  • the transfections are carried out in 24-well plates and each transfection is carried out three times. Twenty four hours before transfection, the cells are seeded: (i) at 5 ⁇ 10 4 cells per well for rabbit smooth muscle cells, cells ECV304, NIH 3T3 and HeLa, (ii) at 10 5 cells per well for cells TU182, (iii) at 3 x 10 4 cells per well for C2C12 cells, and (iv) at 2 x 10 5 cells per well for 293 cells.
  • plasmid DNA 250 ng of plasmid of interest and 250 ng of pUC18
  • DMEM TM medium 20 ⁇ l final
  • the 20 ⁇ l of the DNA / lipid mixture are brought into contact with the cells, in the absence of FCS, for 2 hours.
  • the culture medium is then supplemented with SVF so as to obtain the percentage of SVF required for the culture of each cell type.
  • the culture medium is removed and the cells are rinsed twice with PBS (Life Technologies Inc.).
  • the luciferase activity is then determined using the Luciferase Assay System TM kit (Promega Corporation) according to the supplier's recommendations.
  • the relative activity in rabbit SMC is at least 5 times greater than that observed in other cell types: (i) from 5 to 20 times greater for the hSMact promoter (Table I), and (ii) 5 to 25 times higher for the mSM22 promoter (Table II).
  • the four hybrid promoters therefore possess, in vitro, an activity in smooth muscle cells as important as that of the CMV promoter (which is reputed to be a strong promoter), while retaining a tissue specificity comparable to, or even superior to, that of the promoters.
  • CMV promoter which is reputed to be a strong promoter
  • This example illustrates the tissue specificity properties of the hybrid promoters of the invention in vivo. 3.1. Gene transfer in skeletal muscle.
  • the various plasmids were injected, intramuscularly, into the cranial tibial muscle of female C57BL6 mice aged 5 weeks. Each plasmid, diluted in a NaCl solution at 150 mM final, is injected at a rate of 10 ⁇ g per muscle. Three days after injection, the muscles are removed in 2 ml of Cell Culture Lysis Reagent TM buffer (Promega Corporation), and ground using a Diax homogenizer (Heidolph). The ground material is then centrifuged for 15 minutes at 4000 g, then the luciferase activity is evaluated using the Luciferase Assay System TM kit (Promega Corporation) according to the recommendations of the supplier.
  • Cell Culture Lysis Reagent TM buffer Promega Corporation
  • Diax homogenizer Heidolph
  • the relative activities of the two specific promoters (hSMact and mSM22) as well as those of two of the hybrid promoters of the invention (Enh-hSMact and Enh- mSM22) were also evaluated in vivo after transfer of naked DNA into the cranial tibial muscle of mice.
  • the results collated in FIG. 5 show that the activity of the Enh-hSMact promoter is 100 times lower than that of the CMV promoter.
  • the activity of the Enh-mSM22 promoter is 17 times lower than that of the CMV promoter.
  • EXAMPLE 4 Construction of recombinant adenoviruses expressing the GAX protein under the control of specific hybrid promoters.
  • the purpose of this example is to describe an adenoviral vector carrying the gene coding for the protein GAX operatively linked to the hybrid promoter of the invention composed of the enhancer CMV and of the promoter SM ⁇ -actin (enh-hSMact).
  • the human gax gene comprises 912 base pairs and codes for a transcription factor of 303 amino acids involved in stopping cell growth (growth-arrest-specific homeobox) and having a role in the proliferation of human smooth muscle cells.
  • This homeodomain gene was originally isolated from the aorta and is expressed in particular in adult cardiovascular tissue (Gorski et al. 1993).
  • the sequence of the human gax gene was cloned from a skeletal muscle cDNA library by PCR (Polymerase Chain Reaction) using as a primer a sequence derived from the human gax gene and published by Walsh et al. (Genomics (1994), 24, p535). The sequence was then cloned into the expression vector pXL3297.
  • This plasmid is derived from the Bluescript plasmid (Stratagene) containing the human CMV IE enhancer / promoter (-522 / + 72) (Cell (1985), 41, p521) and the poly A of SV 40 (2538-2759) (GenBank locus SV4CG).
  • Plasmid pXL3297 is an expression vector containing the human gax gene. It was digested with the enzymes HindIII and Avril in order to introduce the human gax gene into the previous plasmid pXL3282 also digested with the enzymes HindIII and Avril to give the plasmid pXL 3300.
  • the final plasmid, pXL3310 comprises an expression cassette consisting of the enhancer CMV-IE, the SM ⁇ -actin promoter (pSMA), associated according to the invention and operationally linked to the gene encoding the human GAX protein as well as the poly A termination signal of SV40.
  • the enhancer CMV-IE the enhancer CMV-IE
  • pSMA SM ⁇ -actin promoter
  • the expression cassette for the human gax gene is then introduced into a recombinant human adenovirus of serotype 5 (Ad5) deleted from the El and E3 regions by co-transfection and homologous recombination between the plasmid carrying the expression cassette for the gax gene and adenovirus, in packaging cells. These cells are preferably line 293.
  • the production of a stock of adenovirus containing the expression cassette for the human gax gene results from the lysis of the cells. packaging 2 or 3 days after infection and isolation of the recombinant viral particles by cesium chloride gradient centrifugation.
  • the viral particles are then used to study the expression of the human gax gene under the control of the promoter of the invention in smooth muscle cells.
  • the expression of the GAX protein is checked 24 hours after infection of the smooth muscle primary cells by immunofluorescence or by western blot using the anti-rabbit polyclonal antibodies.
  • messenger RNAs is analyzed 24 hours after infection of the smooth muscle cells by dot blot and northern blot using an oligonucleotide whose sequence is present in the gax gene.
  • Smooth muscle cells in the exponential growth phase are infected with an adenovirus containing the gene coding for the GAX protein under the control of the enh-hSMact promoter in the absence and in the presence of 125 ng of lipofectin (48-well plates).
  • the adenoviruses (in variable dilutions) and the lipofectamine are incubated for 30 minutes at room temperature in a medium deprived of serum.
  • the mixture or the virus alone is brought into contact with the cells for one hour at 37 ° C.
  • the medium containing the virus is removed and the cells are incubated in DMEM medium containing 0.5% of FCS.
  • the culture medium is replaced by a growth medium for half of the cultures and the incubation is continued for 48 hours to allow the cells to enter the S phase.
  • a weakly mitogenic medium is added to keep the cells in quiescence. Viable cells are counted 72 hours after infection using the Alamar protocol.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Toxicology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
PCT/FR1999/002265 1998-09-25 1999-09-23 Utilisation de promoteurs specifiques hybrides pour controler l'expression tissulaire WO2000018908A1 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CA002343922A CA2343922A1 (fr) 1998-09-25 1999-09-23 Utilisation de promoteurs specifiques hybrides pour controler l'expression tissulaire
HU0105230A HUP0105230A3 (en) 1998-09-25 1999-09-23 Use of specific hybrid promoters for controlling tissue expression
AU56324/99A AU775717B2 (en) 1998-09-25 1999-09-23 Use of specific hybrid promoters for controlling tissue expression
IL14210799A IL142107A0 (en) 1998-09-25 1999-09-23 Use of specific hybrid promoters for controlling tissue expression
JP2000572355A JP2002525109A (ja) 1998-09-25 1999-09-23 組織発現を調節するための特異的ハイブリッドプロモーターの使用
EP99943036A EP1115857A1 (fr) 1998-09-25 1999-09-23 Utilisation de promoteurs specifiques hybrides pour controler l'expression tissulaire
KR1020017003768A KR20010075338A (ko) 1998-09-25 1999-09-23 조직 발현을 제어하기 위한 특정 하이브리드 프로모터의용도

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
FR9812000A FR2783839B1 (fr) 1998-09-25 1998-09-25 Utilisation de promoteurs specifiques hybrides pour controler l'expression tissulaire
FR98/12000 1998-09-25
US12329899P 1999-03-04 1999-03-04
US60/123,298 1999-03-04

Publications (1)

Publication Number Publication Date
WO2000018908A1 true WO2000018908A1 (fr) 2000-04-06

Family

ID=26234566

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/FR1999/002265 WO2000018908A1 (fr) 1998-09-25 1999-09-23 Utilisation de promoteurs specifiques hybrides pour controler l'expression tissulaire

Country Status (9)

Country Link
EP (1) EP1115857A1 (xx)
JP (1) JP2002525109A (xx)
KR (1) KR20010075338A (xx)
CN (1) CN1326506A (xx)
AU (1) AU775717B2 (xx)
CA (1) CA2343922A1 (xx)
HU (1) HUP0105230A3 (xx)
IL (1) IL142107A0 (xx)
WO (1) WO2000018908A1 (xx)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002002765A2 (en) * 2000-07-05 2002-01-10 Transgene S.A. Chimeric promoters for controlling expression in smooth muscle cells
EP1310561A1 (en) * 2001-11-09 2003-05-14 Transgene S.A. Chimeric promoters for controlling expression in skeletal muscle cells

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992017581A1 (en) * 1991-03-28 1992-10-15 Regeneron Pharmaceuticals, Inc. Mammalian expression vector
WO1993009236A1 (en) * 1991-11-06 1993-05-13 Baylor College Of Medicine Myogenic vector systems
WO1996013597A2 (en) * 1994-10-28 1996-05-09 The Trustees Of The University Of Pennsylvania Improved adenovirus and methods of use thereof
WO1998024922A1 (en) * 1996-12-02 1998-06-11 Valentis, Inc. Insulin-like growth factor i (igf-i) expression system and methods of use

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992017581A1 (en) * 1991-03-28 1992-10-15 Regeneron Pharmaceuticals, Inc. Mammalian expression vector
WO1993009236A1 (en) * 1991-11-06 1993-05-13 Baylor College Of Medicine Myogenic vector systems
WO1996013597A2 (en) * 1994-10-28 1996-05-09 The Trustees Of The University Of Pennsylvania Improved adenovirus and methods of use thereof
WO1998024922A1 (en) * 1996-12-02 1998-06-11 Valentis, Inc. Insulin-like growth factor i (igf-i) expression system and methods of use

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
LI L ET AL: "EXPRESSION OF THE SM22ALPHA PROMOTER IN TRANSGENIC MICE PROVIDES EVIDENCE FOR DISTINCT TRANSCRIPTIONAL REGULATORY PROGRAMS IN VASCULAR AND VISCERAL SMOOTH MUSCLE CELLS", THE JOURNAL OF CELL BIOLOGY, vol. 132, no. 5, 1 March 1996 (1996-03-01), pages 849 - 859, XP000609076 *
SHIMIZU ET AL.: "The smooth muscle alpha-actin gene promoter is differentially regulated in smooth muscle versusnon-smooth muscle cells", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 270, no. 13, 31 March 1995 (1995-03-31), pages 7631 - 7643, XP002105507 *
WANG J ET AL: "TARGETED OVEREXPRESSION OF IGF-I EVOKES DISTINCT PATTERNS OF ORGAN REMODELING IN SMOOTH MUSCLE CELL TISSUE BEDS OF TRANSGENIC MICE", JOURNAL OF CLINICAL INVESTIGATION, vol. 100, no. 6, 15 September 1997 (1997-09-15), pages 1425 - 1439, XP002060955 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002002765A2 (en) * 2000-07-05 2002-01-10 Transgene S.A. Chimeric promoters for controlling expression in smooth muscle cells
WO2002002765A3 (en) * 2000-07-05 2002-09-19 Transgene Sa Chimeric promoters for controlling expression in smooth muscle cells
US7482155B1 (en) 2000-07-05 2009-01-27 Transgene S.A. Chimeric promoters for controlling expression in smooth muscle cells
EP1310561A1 (en) * 2001-11-09 2003-05-14 Transgene S.A. Chimeric promoters for controlling expression in skeletal muscle cells

Also Published As

Publication number Publication date
AU775717B2 (en) 2004-08-12
EP1115857A1 (fr) 2001-07-18
JP2002525109A (ja) 2002-08-13
IL142107A0 (en) 2002-03-10
HUP0105230A2 (hu) 2002-04-29
AU5632499A (en) 2000-04-17
HUP0105230A3 (en) 2003-10-28
CN1326506A (zh) 2001-12-12
CA2343922A1 (fr) 2000-04-06
KR20010075338A (ko) 2001-08-09

Similar Documents

Publication Publication Date Title
EP0667912B1 (fr) Vecteurs adenoviraux defectifs et utilisation en therapie genique
KR101752910B1 (ko) 종양 선택적 e1a 및 e1b 돌연변이
EP0698108B2 (fr) Vecteurs adenoviraux d'origine animale et utilisation en therapie genique
WO1995014102A1 (fr) Adenovirus recombinants codant pour la thymidine kinase lors de therapie genique
KR20100105630A (ko) 유인원 아과 c 아데노바이러스 sadv-40, -31, 및 -34 및 그것의 사용
FR2707664A1 (fr) Vecteurs viraux et utilisation en thérapie génique.
FR2712602A1 (fr) Virus recombinants, préparation et utilisation en thérapie génique.
JPH11503011A (ja) 条件発現系
EP1078094B1 (fr) Methodes et compositions pour la production de particules virales
EP1002120B1 (fr) Vecteurs adenoviraux chimeres
FR2782732A1 (fr) Systeme d'expression inductible
FR2755699A1 (fr) Nouvelles constructions et vecteurs pour l'expression ciblee et inductible des genes
FR2762615A1 (fr) Nouveau site interne d'entree des ribosomes et vecteur le contenant
WO2000018908A1 (fr) Utilisation de promoteurs specifiques hybrides pour controler l'expression tissulaire
EP1129204B1 (fr) Nouveau systeme de regulation de l'expression d'un transgene
EP0796338B1 (fr) Lignees cellulaires d'encapsidation pour la transcomplementation de vecteurs retroviraux defectifs
FR2783839A1 (fr) Utilisation de promoteurs specifiques hybrides pour controler l'expression tissulaire
Ehsan et al. Antisense and gene therapy to prevent restenosis
Rubanyi Gene therapy—basic principles and the road from bench to bedside
MXPA01003065A (en) Use of specific hybrid promoters for controlling tissue expression
FR2786198A1 (fr) Nouveau systeme de regulation de l'expression d'un transgene
FR2737221A1 (fr) Nouveaux vecteurs viraux pour la therapie genique

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 99811334.4

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AU BA BB BG BR CA CN CR CU CZ DM EE GD GE HR HU ID IL IN IS JP KP KR LC LK LR LT LV MG MK MN MX NO NZ PL RO RU SG SI SK SL TR TT UA US UZ VN YU ZA

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 56324/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 142107

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2000 572355

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2343922

Country of ref document: CA

Ref document number: 2343922

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/2001/003065

Country of ref document: MX

Ref document number: 1020017003768

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1999943036

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 09787995

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 1999943036

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020017003768

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 56324/99

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1999943036

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: 1020017003768

Country of ref document: KR