EP0944717A1 - Procede de production d'adenovirus recombinants - Google Patents

Procede de production d'adenovirus recombinants

Info

Publication number
EP0944717A1
EP0944717A1 EP97930560A EP97930560A EP0944717A1 EP 0944717 A1 EP0944717 A1 EP 0944717A1 EP 97930560 A EP97930560 A EP 97930560A EP 97930560 A EP97930560 A EP 97930560A EP 0944717 A1 EP0944717 A1 EP 0944717A1
Authority
EP
European Patent Office
Prior art keywords
virus
adenovirus
viruses
supernatant
chromatography
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.)
Ceased
Application number
EP97930560A
Other languages
German (de)
English (en)
French (fr)
Inventor
Francis Blanche
Jean-Marc Guillaume
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.)
Centelion SAS
Original Assignee
Rhone Poulenc Rorer SA
Aventis Pharma SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from FR9608164A external-priority patent/FR2750433B1/fr
Application filed by Rhone Poulenc Rorer SA, Aventis Pharma SA filed Critical Rhone Poulenc Rorer SA
Publication of EP0944717A1 publication Critical patent/EP0944717A1/fr
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • C12N7/02Recovery or purification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/36Selective adsorption, e.g. chromatography characterised by the separation mechanism involving ionic interaction
    • B01D15/361Ion-exchange
    • B01D15/363Anion-exchange
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D61/00Processes of separation using semi-permeable membranes, e.g. dialysis, osmosis or ultrafiltration; Apparatus, accessories or auxiliary operations specially adapted therefor
    • B01D61/14Ultrafiltration; Microfiltration
    • B01D61/145Ultrafiltration
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J41/00Anion exchange; Use of material as anion exchangers; Treatment of material for improving the anion exchange properties
    • B01J41/20Anion exchangers for chromatographic processes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • 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
    • C12N15/861Adenoviral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N2710/10351Methods of production or purification of viral material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography

Definitions

  • the present invention relates to a new process for the production of recombinant adenoviruses. It also relates to purified viral preparations produced by this process.
  • Adenoviruses have certain properties which are particularly advantageous for use as a vector for gene transfer in gene therapy. In particular, they have a fairly broad host spectrum, are capable of infecting quiescent cells, do not integrate into the genome of the infected cell, and have not been associated to date with significant pathologies in man. Adenoviruses have thus been used to transfer genes of interest into the muscle (Ragot et al., Nature 361 (1993) 647), the liver (Jaffe et al., Nature genetics 1 (1992) 372), the nervous system (Akli et al., Nature genetics 3 (1993) 224), etc.
  • Adenoviruses are linear double-stranded DNA viruses approximately 36 (kilobases) kb in size. Their 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, Adl2, etc.) have also been sequenced.
  • adenovirus For their use in gene therapy, different vectors 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).
  • DBP 72kDa DNA binding protein
  • Other vectors include a deletion of another region essential for viral replication and / or spread, 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.
  • the recombinant adenoviruses described in the literature are produced from different adenovirus serotypes. There are in fact different serotypes of adenoviruses, the structure and properties of which vary somewhat, but which 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 adenoviruses CAV2 [manhattan or A26 / 61 strain (ATCC VR-800) for example].
  • Other adenoviruses of animal origin are cited in particular in application WO94 / 26914 incorporated herein by reference.
  • the recombinant adenovirus is a human adenovirus of group C. More preferably, it is an adenovirus Ad2 or Ad5.
  • 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.
  • a 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.
  • One of these lines is for example the 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 transcomplementing recombinant adenoviruses defective for the E1 region, that is to say devoid of all or part of the E1 region, and to produce 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 introducing 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 2 ⁇ to 36 hours). After the cell lysis, the recombinant viral particles are isolated by centrifugation in a cesium chloride gradient.
  • 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. DNA viral can also be introduced in the form of fragments each carrying a part of the recombinant viral genome and a homology zone making it possible, after introduction into the packaging cell, to reconstitute the recombinant viral genome by homologous recombination between the different fragments.
  • a conventional process for the production of adenovirus thus comprises the following steps:
  • the cells for example the 293 cells
  • the incubation then lasts from 40 to 72 hours.
  • the virus is then released from the nucleus by lysis of the cells, generally by several successive thawing cycles.
  • the cell lysate obtained is then centrifuged at low speed (2000 to 4000 rpm) and the supernatant (clarified cell lysate) is then purified by centrifugation in the presence of cesium chloride in two stages:
  • the virus band is in the majority. However, two less dense, thin bands are observed, which observation by electron microscopy has shown that they were empty or broken viral particles for the densest band, and viral subunits (pentons, hexons) for the strip least den.se.
  • the virus is harvested by needle puncture in the centrifuge tube and the cesium is removed by dialysis or desalting.
  • the resolution obtained is average, the authors indicating that virus particles are present in several chromatographic peaks; the yield is low (yield of viral particles: 67%; yield of infectious particles: 49%); and the viral preparation obtained following this chromatographic step is impure.
  • pretreatment of the virus with different enzymes / proteins is necessary.
  • This same article also describes a study of the use of gel permeation chromatography, demonstrating very poor resolution and very low yields (15-20%).
  • the present invention describes a new process for the production of recombinant adenoviruses.
  • the process according to the invention results from modifications of the previous processes at the level of the production phase and / or at the level of the purification phase.
  • the method according to the invention now makes it possible to obtain very quickly and industrially, stocks of viruses in very high quantity and quality.
  • One of the first aspects of the invention relates more particularly to a process for the preparation of recombinant adenoviruses in which the viruses are harvested from the culture supernatant.
  • Another aspect of the invention relates to a process for the preparation of adenovirus comprising an ultrafiltration step.
  • the invention also relates to a method for purifying recombinant adenoviruses comprising a step of anion exchange chromatography.
  • the present invention also describes an improved purification process using gas chromatography. gel permeation, optionally coupled to anion exchange chromatography.
  • the method according to the invention makes it possible to obtain viruses of high quality, in terms of purity, stability, morphology and infectivity, with very high yields and under production conditions fully compatible with industrial requirements and with the regulations concerning the production of therapeutic molecules.
  • the method of the invention uses methods for treating culture supernatants tested on a large scale for recombinant proteins, such as microfiltration or deep filtration, and tangential ultrafiltration. Furthermore, due to the stability of the virus at 37 ° C., this process allows better organization at the industrial stage since, unlike the intracellular method, the harvest time does not need to be precise at half day close. In addition, it guarantees a maximum harvest of the virus, which is particularly important in the case of defective viruses in several regions. On the other hand, the process of the invention allows easier and more precise monitoring of the production kinetics, directly on homogeneous samples of supernatant, without pretreatment, which allows better reproducibility of the productions.
  • the method according to the invention also makes it possible to dispense with the step of lysis of the cells.
  • Cell lysis has several drawbacks. Thus, it can be difficult to envisage breaking cells by freezing-thawing cycles on an industrial level.
  • alternative lysis methods (Dounce, X-press, sonication, mechanical shearing, etc.) have drawbacks: they are potentially aerosol generators difficult to confine for L2 or L3 viruses (level of virus confinement, dependent their pathogenicity or their mode of dissemination), these viruses also tend to be infectious by air; they generate shear forces and / or release of heat which are difficult to control and which decrease the activity of the preparations.
  • the solution of using detergents to lyse the cells would need to be validated and would also require validation of the detergent removal.
  • cell lysis leads to the presence in the medium of numerous cellular debris, which makes the purification.
  • the method of the invention potentially allows better maturation of the virus leading to a more homogeneous population.
  • premature lysis of cells potentially releases empty particles which, although not replicative, are a priori infectious and capable of p. participate in the toxic effect of the virus and increase the specific activity ratio of the preparations obtained.
  • the specific infectivity ratio of a preparation is defined as the ratio of the total number of viral particles, measured by biochemical methods (DO260nm, HPLC, PCR, immunoenzymatic methods, etc.) over the number of viral particles generating an effect biological (formation of lysis plaques on cells in culture in solid medium, transduction of cells).
  • this ratio is determined by making the ratio of the concentration of the particles measured by DO at 260 nm to the concentration of units forming the range of the preparation. This ratio must be less than 100.
  • the results obtained show that the process of the invention makes it possible to obtain a virus of a purity at least equal to its counterpart purified by centrifugation in cesium chloride gradient, in a single step and without prior treatment, starting from a concentrated viral supernatant.
  • a first object of the invention therefore relates to a process for the production of recombinant adenoviruses characterized in that the viral DNA is introduced into a culture of packaging cells and the viruses produced are harvested after release into the culture supernatant.
  • the viruses are harvested following a premature cell lysis carried out mechanically or chemically
  • the cells are not lysed by the intervention of an external factor.
  • the culture is continued for a longer period of time, and the viruses are harvested directly from the supernatant, after spontaneous release by the packaging cells.
  • the virus according to the invention is thus recovered in the cell supernatant, whereas in the previous methods, it is an intracellular virus, more particularly intranuclear.
  • the method according to the invention makes it possible to generate viral particles in high quantity and of better quality.
  • this process makes it possible to avoid the lysis steps which are heavy at the industrial level and generate numerous impurities.
  • the principle of the process is therefore based on the collection of the viruses released in the supernatant. This process may involve a longer culture time than previous techniques based on cell lysis. As indicated above, the harvest time does not have to be precise to the nearest half day. It is essentially determined by the kinetics of release of the viruses into the culture supernatant.
  • the kinetics of virus release can be followed in different ways.
  • analysis methods such as RP-HPLC, IE-HPLC, semi-quantitative PCR (example 4.3), staining of dead cells with trypan blue, measuring the release.
  • LDH-type intracellular enzymes measurement of particles in the supernatant by Coulter-type devices or by light diffraction, immunological (ELISA, RIA, etc.) or nephelometric methods, titration by aggregation in the presence of antibodies , etc.
  • the harvest is carried out when at least 50% of the viruses have been released into the supernatant.
  • the time when 50% of the viruses have been released can be easily determined by carrying out kinetics according to the methods described above. Even more preferably, the harvest is carried out when at least 70% of the viruses have been released into the supernatant. It is particularly preferred to carry out the harvest when at least 90% of the viruses have been released into the supernatant, that is to say when the kinetics reach a plateau.
  • the kinetics of virus release are essentially based on the replication cycle of the adenovirus, and can be influenced by certain factors. In particular, it can vary according to the type of virus used, and in particular according to the type of deletion carried out in the recombinant viral genome.
  • the deletion of the E3 region appears to delay the release of the virus.
  • the virus can be harvested 24-48 hours after infection.
  • a longer culture time seems necessary.
  • the applicant has carried out experiments on the kinetics of release of an adenovirus deficient for the E1 and E3 regions in the cell supernatant, and has shown that the release begins 4 to 5 days approximately post-infection, and lasts until on day 14 approximately. Release generally reaches a plateau between day 8 and day 14, and the titer remains stable for at least 20 days post-infection.
  • the cells are cultured for a period of between 2 and 14 days.
  • the release of the virus can be induced by expression in the packaging cell of a protein, for example viral, involved in the release of the virus.
  • the release can be modulated by expression of the Death protein encoded by the E3 region of the adenovirus (protein E3-11.6K), optionally expressed under the control of an inducible promoter. Therefore, it is possible to reduce the time of release of the viruses and to collect in the culture supernatant, more than 50% of the viruses 24-48 hours post-infection.
  • the culture supernatant is advantageously filtered beforehand.
  • the adenovirus having a size of approximately 0.1 ⁇ m (120 nm) the filtration is carried out by means of membranes having a porosity sufficiently large to allow the virus to pass, but sufficiently fine to retain the contaminants.
  • the filtration is carried out by means of membranes having a porosity greater than 0.2 ⁇ m.
  • the filtration is carried out by successive filtrations on membranes of decreasing porosity. Particularly good results have been obtained by carrying out filtration on depth filters with decreasing porosity lO ⁇ m, l.O ⁇ m then 0.8-0.2 ⁇ m.
  • the filtration is carried out by tangential microfiltration on flat membranes or hollow fibers. More particularly, Millipore flat membranes or hollow fibers having a porosity of between 0.2 and 0.6 ⁇ m can be used. The results presented in the examples show that this filtration step has a yield of 100% (no loss of virus was observed by retention on the filter having the lowest porosity).
  • the applicant has now developed a method for harvesting and purifying the virus from the supernatant.
  • the supernatant thus filtered (or clarified) is subjected to ultrafiltration.
  • This ultrafiltration makes it possible (i) to concentrate the supernatant, the volumes involved being significant, (ii) to carry out a first purification of the virus and (iii) to adjust the buffer of the preparation to the subsequent stages of preparation.
  • the supernatant is subjected to a tangential ultrafiltration.
  • Tangential ultrafiltration consists in concentrating and fractionating a solution between two compartments, retentate and filtrate, separated by membranes of determined cutoff thresholds, by carrying out a flow in the retentate compartment of the device and by applying transmenbranary pressure between this compartment and the filtrate compartment.
  • the flow is generally carried out by means of a pump in the retentate compartment of the device and the transbranar pressure is controlled by means of a valve on the liquid stream of the retentate circuit or of a variable flow pump on the liquid stream of the circuit filtrate.
  • the speed of the flow and the transmembrane pressure are chosen so as to generate few shear forces (number of reynolds less than 5000 sec "1 , preferably less than 3000 sec " 1 , pressure less than 1.0 bar) while avoiding clogging of the membranes.
  • Different systems can be used to carry out ultrafiltration, such as, for example, spiral membranes (Millipore, Amicon), flat membranes or hollow fibers (Amicon, Millipore, Sartorius, Pall, GF, Sepracor).
  • the adenovirus having a mass of approximately 1000 kDa, membranes having a cutoff threshold of less than 1000 kDa, preferably between 100 kDa and 1000 kDa, are advantageously used in the context of the invention.
  • membranes having a cutoff threshold of 1000 kDa or higher in fact leads to a significant loss of virus at this stage.
  • membranes with a cutoff threshold of between 200 and 600 kDa are used, even more preferably between 300 and 500 kDa.
  • the experiments presented in the examples show that the use of a membrane having a cutoff threshold of 300 kDa allows the retention of more than 90% of viral particles while eliminating environmental contaminants (DNA, environmental proteins, cellular proteins, etc.).
  • the use of a cutoff threshold of 500 kDa offers the same advantages.
  • This ultrafiltration step thus constitutes an additional purification compared to the conventional scheme insofar as the contaminants of mass below the cut-off threshold (300 or 500 kDa) are eliminated at least in part.
  • the improvement in the quality of the viral preparation is clear when the aspect of separation is compared after the first ultracentrifugation step according to the two methods.
  • the tube of the viral preparation has a cloudy appearance with a coagulum (lipids, proteins) sometimes coming to touch the band of virus, while in the method of the invention, the preparation after release and ultrafiltration presents an already well resolved band of environmental contaminants which persist in the upper phase.
  • the improvement in quality is also demonstrated when the profiles on ion exchange chromatography of a virus obtained by cell lysis are compared with the virus obtained by ultrafiltration as described in the present invention. Furthermore, it is possible to further improve the quality by continuing the ultrafiltration by a diafiltration of the concentrate. This diafiltration is carried out on the same principle as tangential ultrafiltration, and makes it possible to more completely remove contaminants of size greater than the cut-off threshold of the membrane, while achieving equilibration of the concentrate in the purification buffer.
  • the method according to the invention is therefore characterized in that the viruses are harvested by ultrafiltration.
  • the resulting concentrate can be used directly for purifying the virus.
  • This purification can be carried out by prior conventional techniques, such as centrifugation in a cesium chloride gradient or other ultracentrifugation medium making it possible to separate the particles according to their size, density or coefficient of sedimentation.
  • the results presented in Example 4 indeed show that the virus thus obtained exhibits remarkable characteristics.
  • the viruses are therefore harvested and purified by anion exchange chromatography.
  • cellulose For anion exchange chromatography, different types of support can be used, such as cellulose, agarose (Sepharose gels), dextran (Sephadex gels), acrylamide (Sephacryl gels, Trisacryl gels), silica (TSK-gel SW gels), poly [styrene-divinylbenzene] (Source gels or Poros gels), the copolymer ethylene glycol-methacrylate (Toyopearl HW and TSK-gel PW gels), or mixtures (agarose-dextran: Superdex gel).
  • supports in the form of beads, having the following characteristics: - as spherical as possible,
  • the support is chosen from agarose, dextran, acrylamide, silica, poly [styrene-divinylbenzene], the ethylene glycol-methacrylate copolymer, alone or as a mixture.
  • the support used must be functionalized, by grafting a group capable of interacting with an anionic molecule.
  • the group consists of an ain which can be ternary or quaternary.
  • a ternary amine such as DEAE for example, a weak anion exchanger is obtained.
  • a quaternary amine By using a quaternary amine, a strong anion exchanger is obtained.
  • a strong anion exchanger In the context of the present invention, it is particularly advantageous to use a strong anion exchanger.
  • a chromatography support as used above is used, functionalized by quaternary amines.
  • examples that may be mentioned are Source Q, Mono Q, Q Sepharose, Poros HQ and Poros QE resins, Fractogel TMAE type resins, and Toyopearl Super Q resins.
  • Preferred examples of resins usable in the context of the invention are the
  • Source in particular Source Q, such as Q (Pharmacia), MonoBeads, such as Q (Pharmacia), resins of the Poros HQ and Poros QE type.
  • the MonoBeads support (ball diameter 10 ⁇ 0.5 ⁇ m) has been commercially available for more than 10 years and the Source type resins (15 ⁇ m) or Poros (10 ⁇ m or 20 ⁇ m) for approximately 5 years.
  • These last two supports have the advantage of having a very wide distribution of internal pores (they range from 20 nm to 1 ⁇ m), thus allowing the passage of very large objects through the beads.
  • they offer very little resistance to the circulation of liquid through the gel (therefore very little pressure) and are very rigid. The transport of solutes to the functional groups with which they will interact is therefore very rapid.
  • the Applicant has shown that these parameters are particularly important in the case of the adenovirus, the diffusion of which is slow due to its size.
  • results presented in the examples show that the adenovirus can be purified from the concentrate in a single step of anion exchange chromatography, that the purification yield is excellent (140% in terms of tdu, compared to the 49% value reported by Huyghes et al.) and that the resolution is excellent.
  • the results presented show that the adenovirus obtained has a high infectivity, and therefore has the characteristics required for therapeutic use.
  • Particularly advantageous results have been obtained with a strong anion exchanger, that is to say functionalized with quaternary amines, and in particular with Source Q resin.
  • Source Q15 resin is particularly preferred.
  • another subject of the invention relates to a process for the purification of recombinant adenoviruses from a biological medium, characterized in that it comprises a purification step by strong anion exchange chromatography.
  • the biological medium can be a supernatant of packaging cells producing said virus, a lysate of packaging cells producing said virus, or a prepurified solution of said virus.
  • the chromatography is carried out on a support functionalized with a quaternary amine.
  • the support is chosen from agarose, dextran, acrylamide, silica, poly [styrene-divinylbenzene], ethylene glycol-methacrylate copolymer, alone or as a mixture.
  • a particularly advantageous embodiment is characterized in that the chromatography is carried out on a Source Q resin, preferably Q15.
  • the method described above is advantageously carried out using a supernatant of producer cells, and comprises a prior step of ultrafiltration.
  • This step is advantageously carried out under the conditions defined above, and in particular, it is a tangential ultrafiltration on a membrane having a cutoff threshold of between 300 and 500 kDa.
  • the viruses are harvested and purified by gel permeation chromatography.
  • the gel permeation can be carried out directly on the supernatant, on the concentrate, or on the virus resulting from anion exchange chromatography.
  • the supports mentioned for the anion exchange chromatography can be used in this step, but without functionalization.
  • the preferred supports are agarose (Sepharose gels), dextran (Sephadex gels), acrylamide (Sephacryl gels, Trisacryl gels), silica (TSK-gel gels
  • a preferred method according to the invention therefore comprises an ultrafiltration followed by anion exchange chromatography.
  • Another preferred method includes ultrafiltration followed by anion exchange chromatography, followed by gel permeation chromatography.
  • Another variant of the invention relates to a method for purifying adenovirus from a biological medium comprising a first stage of ultracentrifugation, a second stage of dilution or dialysis, and a third stage of anion exchange chromatography .
  • the first step is carried out by rapid ultracentrifugation on a cesium chloride gradient.
  • rapid means ultracentrifugation ranging from about 0.5 to 4 hours.
  • the virus is diluted or dialyzed against buffer, to facilitate its injection on the chromatography gel, and the elimination of the ultracentrifugation medium.
  • the third step is carried out using anion exchange chromatography as described above, preferably strong anions.
  • a 1st rapid ultracentrifugation is carried out with cesium chloride (as in Example 3). Then, after a simple dilution of the sample (for example by 10 volumes of buffer) or after a simple dialysis in buffer, the sample is chromatographed in ion exchange (as in example 5.1.).
  • the advantage of this variant of the process of the invention comes from the fact that it implements 2 completely different modes of separation of the virus (density and surface charge), which can possibly bring the virus to a level of quality combining the performances of the 2 methods.
  • the chromatography step allows simultaneously to eliminate the medium used for ultracentrifugation (cesium chloride for example, or any other equivalent medium mentioned above).
  • Another subject of the invention relates to the use of iodixanol, 5.5 ′ - [(2-hydroxy-1-3-propanediyl) -bis (acetylamino)] bis [N, N'-bis (2,3dihydroxypropyl-2, 4,6-triodo- 1, 3-benzenecarboxamide] for the purification of adenovirus.
  • the packaging cells can be prepared from different pharmaceutically usable cells, that is to say cultivable under industrially acceptable conditions and having no recognized pathogenic character. They can be established cell lines or primary cultures and in particular human retinoblasts, human lung carcinoma cells, or kidney embryonic cells. Advantageously, these are cells of human origin, which can be infected with an adenovirus. In this regard, mention may be made of KB, Hela, 293, Vero, gmDBP6, HER, A549, HER, etc. cells.
  • the cells of the KB line are derived from a human epidermal carcinoma. They are accessible to ATCC (ref. CCL17) as well as the conditions allowing their cultivation.
  • the Hela human cell line comes from a carcinoma of the human epithelium. It is also accessible to ATCC (ref. CCL2) as well as the conditions allowing its cultivation.
  • the cells of line 293 are human embryonic kidney cells (Graham et al., J. Gen. Virol. 36 (1977) 59). This line contains in particular, integrated into its genome, the left part of the genome of the human adenovirus Ad5 (12%).
  • the gm DBP6 cell line (Brough et al., Virology 190 (1992) 624) consists of Hela cells carrying the E2 adenovirus gene under the control of the LTR of MMTV.
  • MDCK canine origin
  • cells from the MDCK canine line are preferred. Culture conditions MDCK cells have been described in particular by Macatney et al., Science 44 (1988) 9.
  • Different packaging cell lines have been described in the literature and are mentioned in the examples. They are advantageously cells trans-complementing the El function of the adenovirus. Even more preferably, these are cells that complement the E1 and E4 or E1 and E2a functions of the adenovirus. These cells are preferably derived from human embryonic kidney or retinal cells, or from human lung carcinomas.
  • the invention thus provides a method of producing particularly advantageous recombinant adenoviruses.
  • This method is suitable for the production of recombinant viruses defective for one or more regions, and in particular, viruses defective for the El region, or for the El and E4 regions. Furthermore, it is applicable to the production of adenoviruses of different serotypes, as indicated above.
  • the method of the invention is used for the production of recombinant adenoviruses in which the E1 region is inactivated by deletion of a PvuII-BglII fragment going from nucleotide 454 to nucleotide 3328, on the sequence of l adenovirus Ad5.
  • This sequence is accessible in the literature and also on the database (see in particular Genebank n ° M73260).
  • the E1 region is inactivated by deletion of a HinfII-Sau3A fragment going from nucleotide 382 to nucleotide 3446.
  • the method allows the production of vectors comprising a deletion of the entire region E4.
  • E4 a Maell-Mscl fragment corresponding to nucleotides 35835-32720.
  • a functional part of E4 is deleted. This part includes at least the ORF3 and ORF6 phases.
  • these coding phases can be deleted from the genome in the form of PvuII-AluI and BglII-PvuII fragments respectively, corresponding to nucleotides 34801-34329 and 34115-33126 respectively.
  • the deletions of the E4 region of the Ad2 dl808 virus or of the Ad5 dll004, Ad5 dll007, Ad5 dllOll or Ad5 dll014 viruses can also be used in the context of the invention.
  • the cells of the invention are particularly advantageous for the production of viruses comprising an inactive E1 region and a deletion in the E4 region of the type of that present in the genome of Ad5 d11014, that is to say of E4 virus "retaining the ORF4 reading phase.
  • the deletion in the El region advantageously covers all or part of the El A and E1B regions. This deletion must be sufficient to render the virus incapable of autonomous replication in a cell.
  • the part of the El region deleted in the adenoviruses according to the invention advantageously covers the nucleotides 454-3328 or 382-3446.
  • the adenoviruses produced may have other alterations in their genome.
  • other regions can be deleted to increase the capacity of the virus and reduce its side effects linked to the expression of viral genes.
  • all or part of the E3 or IVa2 region in particular can be deleted.
  • the E3 region it may however be particularly advantageous to keep the part coding for the protein gpl9K.
  • This protein in fact makes it possible to prevent the adenoviral vector from being the subject of an immune reaction which (i) limits its action and (ii) could have undesirable side effects.
  • the E3 region is deleted and the sequence coding for the protein gpl9k is reintroduced under the control of a heterologous promoter.
  • adenoviruses constitute very efficient gene transfer vectors for gene and cell therapy applications.
  • a heterologous nucleic acid sequence whose transfer and / or expression in a cell, an organ or an organism is sought can be inserted into their genome.
  • This sequence may contain one or more therapeutic genes, such as a gene whose transcription and possibly translation into the target cell generate products having a therapeutic effect.
  • therapeutic products mention may more particularly be made of enzymes, blood derivatives, hormones, lymphokines: interleukins, interferons, TNF, etc.
  • FR 9203120 growth factors, neurotransmitters or their synthetic precursors or enzymes, trophic factors: BDNF, CNTF, NGF, IGF, GMF, aFGF, bFGF, NT3, NT5, etc; apolipoproteins: ApoAI, ApoAIV, ApoE, etc. (WO94 / 25073), dystrophin or a minidystrophin (WO93 / 06223), tumor suppressor genes: p53, Rb, RaplA, DCC, k-rev, etc.
  • trophic factors BDNF, CNTF, NGF, IGF, GMF, aFGF, bFGF, NT3, NT5, etc
  • apolipoproteins ApoAI, ApoAIV, ApoE, etc.
  • WO93 / 06223 tumor suppressor genes: p53, Rb, RaplA, DCC, k-rev, etc.
  • WO94 / 24297 genes coding for factors involved in coagulation: Factors VII, VIII, IX, etc., suicide genes: thymidine kinase, cytosine deaminase, etc; or all or part of a natural or artificial immunoglobulin (Fab, ScFv, etc., WO94 / 29446), etc.
  • the therapeutic gene can also be an antisense gene or sequence, the expression of which in the target cell makes it possible to control the expression of genes or the transcription of cellular mRNAs. Such sequences can for example be transcribed, in the target cell, into RNAs complementary to cellular mRNAs and thus block their translation into protein, according to the technique described in patent EP 140 308.
  • the therapeutic gene can also be a gene encoding for an antigenic peptide capable of generating an immune response in humans, for the production of vaccines.
  • antigenic peptides capable of generating an immune response in humans, for the production of vaccines.
  • These may in particular be antigenic peptides specific for the epstein barr virus, the HIV virus, the hepatitis B virus (EP 185 573), the pseudo-rabies virus, or even specific for tumors (EP 259 212).
  • the heterologous nucleic acid sequence also comprises a promoter region for functional transcription in the infected cell, as well as a region located 3 ′ of the gene of interest, and which specifies a transcriptional end signal and a site for polyadenylation. All of these elements constitute the expression cassette.
  • the promoter region it may be a promoter region naturally responsible for the expression of the gene considered when it is capable of functioning in the infected cell. It can also be from regions of different origin (responsible for the expression of other proteins, or even synthetic).
  • they may be promoter sequences of eukaryotic or viral genes or any promoter or derivative sequence, stimulating or repressing the transcription of a gene in a specific way or not and in an inducible way or not.
  • they may be promoter sequences originating from the genome of the cell which it is desired to infect, or from the genome of a virus, and in particular, the promoters of the E1A, MLP genes of adenovirus, the CMV promoter, LTR-RSV, etc.
  • eukaryotic promoters include ubiquitous promoters (HPRT, vimentin, ⁇ -actin, tubulin, etc.), promoters of intermediate filaments (desmin, neurofilaments, keratin, GFAP, etc.) promoters of therapeutic genes (MDR type , CFTR, factor VIII, etc.) tissue-specific promoters (pyruvate kinase, villin, promoter of the intestinal fatty acid binding protein, promoter of actin to smooth muscle cells, specific promoters for the liver; Apo AI , Apo AU, Human albumin, etc.) or promoters responding to a stimulus (steroid hormone receptor, retinoic acid receptor, etc.).
  • HPRT ubiquitous promoters
  • promoters of intermediate filaments desmin, neurofilaments, keratin, GFAP, etc.
  • promoters of therapeutic genes MDR type , CFTR, factor VIII, etc.
  • tissue-specific promoters pyruvate kinase, vi
  • these expression sequences can be modified by adding activation, regulation sequences or allowing tissue-specific or majority expression.
  • the inserted nucleic acid does not contain expression sequences, it can be inserted into the genome of the defective virus downstream of such a sequence.
  • heterologous nucleic acid sequence may also comprise, in particular upstream of the therapeutic gene, a signal sequence directing the therapeutic product synthesized in the secretory pathways of the target cell.
  • This signal sequence may be the natural signal sequence of the therapeutic product, but it may also be any other functional signal sequence, or an artificial signal sequence.
  • the therapeutic gene expression cassette can be inserted at different sites in the genome of the recombinant adenovirus, according to the techniques described in the prior art. It can first of all be inserted at the level of the El deletion. It can also be inserted at the E3 region, in addition to or in substitution for sequences. It can also be located at the level of the deleted E4 region.
  • the present invention also relates to the purified viral preparations obtained according to the process of the invention, as well as any pharmaceutical composition comprising one or more defective recombinant adenoviruses prepared according to this process.
  • 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.
  • pharmaceutically acceptable vehicles for an injectable formulation may in particular be saline solutions (monosodium phosphate, disodium phosphate, sodium chloride, potassium, calcium or magnesium, etc., or mixtures of such salts), sterile, isotonic, or dry compositions, in particular lyophilized, which, by addition, as appropriate, of sterilized water or physiological saline, allow the constitution of injectable solutes.
  • 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 doses of virus used for the injection can be adapted according to various parameters, and in particular according to the mode of administration used, the pathology concerned, the gene to be expressed, or even the duration of the treatment sought.
  • the recombinant adenoviruses according to the invention are formulated and administered in the form of doses between 10 ⁇ and 10 *** ⁇ pfu, and preferably 10 ° to 10 *** ⁇ pfu.
  • pfu plaque forming unit
  • plaque forming unit corresponds to the infectious power of an adenovirus solution, and is determined by infection of an appropriate cell culture, and measures, generally after 15 days, the number of plaques of infected cells.
  • the techniques for determining the pfu titer of a viral solution are well documented in the literature.
  • the viruses thus produced can be used for the treatment or prevention of many pathologies, including genetic diseases (dystrophy, cystic fibrosis, etc.), neurodegenerative diseases (Alzheimer, Parkinson, ALS, etc.), cancers , pathologies linked to coagulation disorders or dyslipoproteinemias, pathologies linked to viral infections (hepatitis, AIDS, etc.), etc.
  • Figure 1 Study of the stability of the purified adenovirus according to Example 4.
  • Figure 2 Analysis in HPLC (reverse phase) of the purified adenovirus according to Example 4. Comparison with the adenovirus of Example 3.
  • Figure 3 Release kinetics of the Ad- ⁇ Gal adenovirus in the supernatant of 293 cells, measured by semi-quantitative PCR and Plate Assay.
  • Figure 4 Elution profile on Source Q15 of an ultrafilter adenovirus supernatant (Example 5.1).
  • FIG. 5 Analysis in HPLC Resource Q of the peak of virus harvested by chromatography on resin Source Q15 of a supernatant of adenovirus ultrafilter (example 5.1).
  • Figure 6 (A) Elution profile on Source Q15 resin of an adenovirus supernatant Ad-APOAl ultrafilter (Example 5.3); and (B) HPLC analysis (Resource Q) of the peak of harvested virus.
  • Figure 7 (A) Elution profile on Source Q15 of an adenovirus supernatant Ad-TK ultrafilter (Example 5.3). HPLC analysis (Resource Q) of the different fractions (beginning and end of peak) of harvested viruses: (B) F2 fraction, middle of the peak; (C) Fraction F3, peak bound; (D) Fraction F4, end of the peak.
  • FIG 8 Elution profile on Mono Q resin of a concentrated supernatant of culture of adenovirus producing cells (example 5.4).
  • BG25F1 Supernatant virus concentrates and purifies on cesium.
  • BG25C Supernatant infects, concentrates.
  • Figure 9 Elution profile on POROS HQ gel of a concentrated supernatant from the culture of adenovirus producing cells (Example 5 4) BG25F1 Supernatant virus concentrated and purified on cesium BG25C Infectant supernatant, concentrated
  • Figure 1 Analysis by electron microscopy of a purified adenovirus stock according to the invention
  • the plasmids of the pBR322, pUC type and the phages of the Ml 3 series are of commercial origin (Bethesda Research Laboratories)
  • the DNA fragments can be separated according to their size by electrophoresis in agarose gels or acrylamide, extracts with phenol or with a phenol / chloroform mixture, precipitated with ethanol then incubated in the presence of phage T4 DNA ligase (Biolabs) according to the supplier's recommendations
  • the filling of the prominent 5 'ends can be carried out by the Klenow fragment of E coli DNA Polymerase I (Biolabs) according to supplier's specifications Destruction of the prominent 3 'ends is carried out in the presence of phage DNA Polymerase T4 (Biolabs) used according to the manufacturer's recommendations.
  • the destruction of the protruding 5 ′ ends is carried out by gentle treatment with nuclease SI.
  • Mutagenesis directed in vitro by synthetic oligodeoxynucleotides can be carried out according to the method developed by Taylor et al. [Nucleic Acids Res. 12 (1985) 8749-8764] using the kit distributed by Amersham.
  • Enzymatic amplification of DNA fragments by the technique called PCR can be performed using a "DNA thermal cycler" (Perkin Elmer Cetus) according to the manufacturer's specifications. Verification of the nucleotide sequences can be carried out by the method developed by Sanger et al. [Proc. Natl. Acad. Sci. USA, 74 (1977) 5463-5467] using the kit distributed by Amersham.
  • Example 1 Cell lines for packaging
  • the packaging cells used in the context of the invention can come from any cell line infectable with an adenovirus, compatible with use for therapeutic purposes. It is more preferably a cell chosen from the following lines:
  • Line 293 is a line of human kidney embryonic cells 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, Elb, the region coding for the pIX protein and part of the region coding for the pIVa2 protein (Graham et al., J. Gen. Virol. 36 (1977) 59 ).
  • Ad5 adenovirus serotype 5
  • This line is capable of trans-complementing recombinant adenoviruses defective for the E1 region, that is to say devoid of all or part of the E1 region, and to produce viral stocks having high titers -
  • the cells of the A549 line are capable of trans-complementing recombinant adenoviruses defective for the E1 region, that is to say devoid of all or part of the E1 region, and to produce viral stocks having high titers -
  • the cells of the A549 line are capable of trans-complementing recombinant adenoviruses defective for the E1 region, that is to say devoid of all or part of the E1 region, and to produce viral stocks having high titers -
  • the cells of the A549 line are capable of trans-complementing recombinant adenoviruses defective for the E1 region, that is to say devoid of all or part of the E1 region, and to produce viral stocks having high titers
  • Cells complementing the E1 region of the adenovirus were constructed from A549 cells (Imler et al., Gene Ther. (1996) 75). These cells contain a fragment of the El region, devoid of the left ITR, placed under the control of an inducible promoter.
  • HER Human retinal embryonic cells
  • adenovirus Bactet al., Oncogene 2 (1988) 477
  • Adenovirus packaging cells prepared from these cells have been described for example in application WO94 / 28152 or in the article by Fallaux et al. (Hum.Gene Ther. (1996) 215).
  • Mention may more particularly be made of the line 911 comprising the E1 region of the genome of the adenovirus Ad5, from nucleotide 79 to nucleotide 5789 integrated into the genome of HER cells. This cell line allows the production of viruses defective for the E1 region.
  • IGRP2 cells are cells obtained from 293 cells, by integration of a functional unit of the E4 region under the control of an inducible promoter. These cells allow the production of viruses defective for the E1 and E4 regions (Yeh et al., J. Virol (1996) 70).
  • VK cells (VK2-20 and VK10-9) are cells obtained from 293 cells, by integration of the entire E4 region under the control of an inducible promoter, and of the region coding for the protein pIX. These cells allow the production of viruses defective for the E1 and E4 regions (Krougliak et al., Hum. Gene Ther. 6 (1995) 1575).
  • the 293E4 cells are cells obtained from 293 cells, by integration of the entire E4 region. These cells allow the production of viruses defective for the El and E4 regions (WO95 / 02697; Cancer Gene Ther. (1995) 322).
  • viruses produced in the context of the following examples are an adenovirus containing the LacZ marker gene from E. coli (Ad- ⁇ Gal), an adenovirus containing the gene coding for the herpes virus type I thymidine kinase (Ad-TK ), an adenovirus containing the gene encoding the human p53 tumor suppressor protein and a virus encoding the apolipoprotein A1 (Ad-apoAI). These viruses are derived from the Ad5 serotype and have the following structure:
  • a deletion in the E1 region covering for example, nucleotides 382 (Hinfl site) to 3446 (Sau3a site).
  • any other construct can be produced according to the method of the invention, and in particular viruses carrying other heterologous genes and / or other deletions (E1 / E4 or E1 / E2 for example).
  • This example reproduces the prior technique of virus production, consisting in lysing the packaging cells to recover the viruses produced.
  • the incubation lasts from 40 to 72 hours, the harvest timing being judged by the observation under the microscope of the cells which round, become more refractive and adhere more and more weakly to the culture support.
  • the kinetics of the viral cycle last from 24 to 36 hours.
  • the concentration factor is depending on the size of the culture of the order of 10 to 100 times.
  • the virus is then released from the nucleus by 3 to 6 successive thawing cycles (dry ice ethanol at -70 ° C, water bath at 37 ° C)
  • the cell lysate obtained is then centrifuged at low speed (2000 at 4000 rpm) and the supernatant (clarified cell lysate) is then purified by ultracentrifugation in cesium chloride gradient in two stages:
  • the rotors can be 'swinging' rotors (Sw28, S 41 Beckman) or fixed angle (Ti 45, Ti 70, Ti 70.1 Beckman) depending on the volumes to be treated;
  • a second ultracentrifugation in a longer gradient (from 10 to 40 hours depending on the rotor used), for example 18 hours at 35,000 rpm in sw 41 rotor which constitutes the true and unique purification of the virus.
  • the virus is found in a linear gradient at equilibrium at a density of 1.34.
  • the virus band is in the majority. Sometimes, however, two less dense, thin bands are observed, which observation by electron microscopy has shown that they were empty or broken viruses and for the less dense band of viral subunits (pentons, hexons). After this stage, the virus is harvested in the tube by needle puncture and the cesium is eliminated by dialysis or desalting on G25.
  • EXAMPLE 4 Production of Viruses in the Supernatant This example describes an experiment in virus production by recovery after spontaneous release. The virus is then harvested by ultrafiltration, then purified by cesium chloride.
  • the cells are not harvested 40 to 72 hours post-infection, but the incubation is prolonged between 8 to 12 days so as to obtain a total lysis of the cells without the need to proceed freezing and thawing cycles.
  • the virus is found in the supernatant.
  • the supernatant is then clarified by filtration through filters with decreasing porosity depth (10 ⁇ m / 1.0 ⁇ m / 0.8-0.2 ⁇ m).
  • the virus has a size of 0.1 ⁇ m and at this stage we did not observe any loss of virus by retention on the filter with the lowest porosity (0.22 ⁇ m)
  • the supernatant once clarified is then concentrated by tangential ultrafiltration on a Millipore spiral membrane having a cutoff threshold of 300 kDa.
  • the concentration factor is dictated by the dead volume of the system which is 100 ml. Volumes of supernatant from 4 to 20 liters were concentrated with this system, making it possible to obtain volumes of concentrate from 100 ml to 200 ml without difficulty, which corresponds to a concentration factor of 20 to 100 times.
  • the concentrate is then filtered through 0.22 ⁇ m and then purified by centrifugation on cesium chloride as described in Example 3, followed by a dialysis step.
  • the intracellular virus tube presents a cloudy appearance with a coagulum (lipids, proteins) sometimes coming to touch the band of virus
  • the viral preparation obtained after the first stage of centrifugation on cesium chloride by the procedure of The invention presents a band of virus already well isolated from the environmental contaminants which persist in the upper phase.
  • the analysis in high performance liquid chromatography on a Resource Q column also shows this gain in purity of the starting material obtained by ultrafiltration of infected supernatant with a reduction in nucleic acid contaminants (DO 260/280 ratio greater than or equal to 1.8) and protein (DO 260/280 ratio less than 1).
  • the stability of the virus was determined by titration by the plate assay method of an infectious culture supernatant from which aliquots were taken at different incubation times at 37 ° C. post-infection. The results are presented below:
  • Ad-TK virus Ad-TK virus
  • This parameter corresponding to the ratio of the number of viral particles measured by HPLC to the number of pfu, accounts for the infectious power of the viral preparations. According to FDA recommendations, it should be less than 100.
  • This parameter was measured as follows: Two series of culture flasks containing 293 cells were infected in parallel at the same time with the same viral prestock under the same conditions. This experiment was carried out for a recombinant Ad-bgal adenovirus, then repeated for an Ad-TK adenovirus. For each adenovirus, a series of vials is harvested 48 hours post-infection and is considered to be a production of purified intracellular virus on a cesium gradient after freezing and thawing.
  • the other series is incubated 10 days post-infection and the virus is harvested from the supernatant.
  • the purified preparations obtained are titrated by assay plate and the quantification of the number of total viral particles is determined by measuring the concentration of PVII protein by reverse phase HPLC on a C4 Vydac 4.6 ⁇ 50 mm column after denaturation of the samples in 6.4M guanidine. .
  • Protocol 20 ⁇ l of sample are placed on a carbon grid and then treated for observation in negative staining with 1.5% uranyl acetate. For observation, a Jeol 1010 electron microscope from 50 kV to 100 kV is used.
  • Figure 2 shows the overlay of 3 chromatograms obtained from two virus samples collected intracellularly and a virus sample purified by the supernatant method.
  • the chromatograms show a perfect identity between the samples, with no difference in the relative intensities of each peak. The nature of each peak was determined by sequencing and demonstrates that the proteins present are all of viral origin (see table below).
  • the cytotoxicity analysis is carried out by infecting HCT116 cells in 24-well plates for increasing MOIs and by determining the percentage of living cells compared to an uninfected control, 2 and 5 days post-infection, using the technique of staining with violet crystal. The results are shown in the table below:
  • the transduction efficiency of a preparation is determined by infecting W162 cells, non-permissive to replication, cultured in 24-well plates, with increasing concentrations of viral particles. For the same quantity of viral particles deposited, there are, 48 hours post-infection, the cells expressing the betagalactosidase activity after incubation with X-gal as substrate. Each blue cell is counted as a transduction unit (TDU), the result is multiplied by the dilution of the sample in order to obtain the concentration in unit of transductions of the sample. The transduction efficiency is then expressed by making the ratio of the concentration of viral particles to the concentration of TDU. The results obtained show that the purified viruses exhibit good transduction efficiency in vitro.
  • TDU transduction unit
  • the adenoviruses were injected stereotaxically into the striatum of OF1 immunocompetent mice. For this, volumes of l ⁇ l to 10 'pfu of virus were injected at the following stereotaxic landmarks (for the 0mm incisor bar): Anteroposterior: +0.5; Medio-lateral: 2; Depth: -3.5.
  • the brains were analyzed 7 days after the injection. The results obtained show that the transduction efficiency is high: thousands of transduced cells, very intense expression in the nucleus and frequent and intense diffusion in the cytoplasm.
  • This example describes a study of the kinetics of release of adenoviruses in the culture supernatant of packaging cells.
  • the amount of adenovirus released into the supernatant was determined on a supernatant of 293 cells infected with Ad- ⁇ Gal, at different post-infection times. The results obtained are shown in Figure 3. They show that cell release begins from the 5th or 6th day post infection.
  • virus determination technique can be used for the same purpose, on any other packaging line, and for any type of adenovirus.
  • EXAMPLE 5 Purification of the Virus by Ultrafiltration and Ion Exchange This example illustrates how the adenovirus contained in the concentrate can be purified directly and in a single chromatographic step of ion exchange, with very high yields.
  • the starting material therefore consists of the concentrate
  • This retentate has a total protein content of between 5 and 50 mg / ml, and more preferably between 10 and 30 mg / ml, in PBS buffer (10 mM phosphate , pH 7.2 containing 150 mM NaCl).
  • the ultrafiltration supernatant obtained from a virus preparation is injected into a column containing Source Q 15 (Pharmacia) equilibrated in 50 mM Tris / HCl buffer pH 8.0 containing 250 mM NaCl, 1.0 mM MgC12, and 10% glycerol (buffer A). After rinsing with 10 column volumes of buffer A, the adsorbed species are eluted with a linear gradient of NaCl (250 mM to 1 M) on 25 column volumes at a linear flow rate of 60 to 300 cm / h, more preferably 12 cm / h. The typical elution profile obtained at 260 nm is presented in FIG. 4. The fraction containing the viral particles is collected.
  • the fraction collected has a purity of 98% in viral particles (UV detection at 260 nm), when it is analyzed by high performance liquid chromatography (HPLC) on a column of Resouce Q (1 ml) in a following chromatographic system: 10 ⁇ l of the fraction purified by chromatography as described in Example 5.1 are injected onto a Resource Q15 column (1 ml of gel; Pharmacia) balanced in 50 mM Tris / HCl buffer pH 8.0 (buffer B) . After rinsing with 5 ml of buffer B, the adsorbed species are eluted with a linear gradient of 30 ml of NaCl (0 to 1 M) in buffer B at a flow rate of 1 ml / min.
  • HPLC high performance liquid chromatography
  • the electrophoresis analysis of the purified adenoviral fraction by chromatography is carried out on polyacrylamide gel (4-20%) under denaturing conditions (SDS). The protein bands are then revealed with silver nitrate.
  • adenoviral preparation obtained by chromatography has a level of purity at least equal to that of the preparation conventionally maintained by ultracentrifugation since it does not have an additional protein band which would indicate contamination of the preparation with non-adenoviral proteins.
  • the adenoviral preparation obtained by chromatography has an absorbance ratio A 2 60 nm At 280 nm equal to 1.30 ⁇ 0.05. This value, which is identical to that obtained for the best preparations obtained by ultracentrifugation, indicates that the preparation is devoid of contaminating proteins or contaminating nucleic acids.
  • Example 4.2 The analysis by electron microscopy carried out under the conditions described in Example 4.2 on an Ad- ⁇ gal virus purified by chromatography shows a clean preparation, without contaminants, without aggregates and without empty viral particles (FIG. 11).
  • the ultracentrifugation in cesium chloride gradient of this preparation reveals a single band of density 1.30, which confirms the absence of contamination of the chromatographic preparations by possible empty particles or fragments of capsids.
  • the chromatographic peak of the virus is followed by a shoulder (or secondary peak) in its rear part, which is not collected with the main peak.
  • the cesium chloride gradient ultracentrifugation of this fraction reveals a density band 1.27, and analysis of the composition of this fraction shows that it does not contain nucleic acids.
  • adenoviruses comprising a therapeutic gene such as the genes coding for the ApoAl proteins or thimidine kinase.
  • This example illustrates how adenoviruses comprising in their genomes heterologous nucleic acid sequences encoding proteins can be purified directly and in a single chromatographic ion exchange step. It also shows that the chromatographic behavior of the adenovirus is independent of the heterologous nucleic acid sequences that it carries, which makes it possible to implement the same purification process for different adenoviruses carrying diverse heterologous nucleic acid sequences.
  • an adenovirus comprising in its genome a heterologous nucleic acid sequence coding for the ApoAl protein (Example 2, WO94 / 25073) is purified by chromatography, in the system described in Example 5.1, 18 ml (72 mg of proteins; 1.08 x 10 3 particles) of concentrated supernatant from a cell culture harvested 10 days post-infection (FIG. 6A).
  • the peak of viral particles collected after chromatography contained 9.98 x 10 12 particles, which indicates a particle yield of 92% and a purification factor of 51.
  • an adenovirus comprising in its genome a heterologous nucleic acid sequence coding for the herpes simplex protein thimidine kinase type 1 (Example 2, WO95 / 14102) is purified by chromatography in the system described in Example 5.1. 36 ml (180 mg protein; 4.69 x 10 13 particles) of concentrated cell culture supernatant (Figure 7A). The peak of viral particles collected after chromatography (20 ml; 5.6 mg of protein) contained 4.28 x 10 n particles, which indicates a particle yield of 91% and a purification factor of 32.
  • This example illustrates how an adenovirus comprising in its genome a heterologous nucleic acid sequence can be purified directly in a single chromatographic step of ion exchange from a lysate of packaging cells producing said virus.
  • an adenovirus comprising in its genome a heterologous nucleic acid sequence coding for the ⁇ -gal protein is purified by chromatography in the system described in example 5.1 (column
  • FineLine Pilot 35, Pharmacia 100 ml of Source 15Q resin), 450 ml (i.e. 2.5 x 10 * 4 particles) of concentrated lysate from a cell culture harvested 3 days post-infection by chemical lysis (1% Tween-20).
  • the peak of viral particles collected after chromatography 110 ml contained 2.15 x 10 4 particles, which indicates a particle yield of 86%. After this purification step, the fraction collected had a purity greater than 98% in viral particles during the chromatographic analysis under the conditions described in 5.2.
  • Example 5.2 The electrophoresis analysis of the purified adenoviral fraction by chromatography carried out under the conditions described in Example 5.2 showed that this preparation had a level of purity at least equal to that of a preparation obtained conventionally by ultracentrifugation, and that it is devoid of contaminating proteins or contaminating nucleic acids. 5.5. Purification of the virus by ultrafiltration and ion exchange chromatography on different columns.
  • This example illustrates how the adenovirus contained in the concentrate can be purified directly and in a single chromatographic ion exchange step using a gel different from the Source 15 Q support, while operating on the same separation principle, anion exchange by interaction with quaternary amino groups in the matrix.
  • Source Q30 gel can therefore be suitable for the purification of recombinant adenoviruses, even if its properties remain inferior to those of Source Q15 (purity on the order of 99%, efficiency on the order of 8000 and ability to 2.5 to 5 10 12 pv per ml).
  • a ⁇ -Gal adenovirus is purified by chromatography on a column of MonoQ HR 5/5 according to the protocol described in Example 5.1.
  • the chromatographic image corresponding to the ultrafiltration retentate and to the purified viral preparation thus obtained is illustrated in FIG. 8.
  • a ⁇ -Gal adenovirus is purified by chromatography on a Poros HQ / M column following the protocol described in Example 5.1.
  • the corresponding chromatographic image to the ultrafiltration retentate and to the purified viral preparation thus obtained is illustrated in FIG. 9.
  • This example illustrates how the adenovirus contained in the concentrate (ultrafiltration retentate) can be purified directly by gel permeation chromatography, with very high yields.
  • the resolution of the two gel permeation chromatographic systems described above can be advantageously improved by chromatographing the ultrafiltration supernatant (200 ⁇ l) on a system of 2 columns HR 10/30 (Pharmacia) coupled in series (Sephacryl S column -1000HR or S-2000 followed by a column of Superdex 200 HR) equilibrated in buffer C.
  • the species are eluted with buffer C at a flow rate of 0.5 ml / min and detected in UV at 260 nm.
  • the peak of viral particles is very clearly better separated from the lower molecular weight species than in the system comprising a Sephacryl S-1000 HR or Sephacryl S-2000 column alone.
  • Example 7 Purification of the virus by Ultrafiltration. Ion exchange and gel permeation
  • the fraction of viral particles resulting from anion exchange chromatography can advantageously be chromatographed in one of the gel permeation chromatographic systems described above, for example in order to further improve the level purity of the viral particles, but also mainly for the purpose of conditioning the viral particles in a compatible or adapted medium for subsequent uses of the viral preparation (injection, etc.).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Water Supply & Treatment (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Plant Pathology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
EP97930560A 1996-07-01 1997-06-20 Procede de production d'adenovirus recombinants Ceased EP0944717A1 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
FR9608164 1996-07-01
FR9608164A FR2750433B1 (fr) 1996-07-01 1996-07-01 Procede de production d'adenovirus recombinants
US2666796P 1996-09-25 1996-09-25
US26667P 1996-09-25
PCT/FR1997/001107 WO1998000524A1 (fr) 1996-07-01 1997-06-20 Procede de production d'adenovirus recombinants

Publications (1)

Publication Number Publication Date
EP0944717A1 true EP0944717A1 (fr) 1999-09-29

Family

ID=26232805

Family Applications (1)

Application Number Title Priority Date Filing Date
EP97930560A Ceased EP0944717A1 (fr) 1996-07-01 1997-06-20 Procede de production d'adenovirus recombinants

Country Status (13)

Country Link
US (3) US6485958B2 (no)
EP (1) EP0944717A1 (no)
JP (1) JP2000514290A (no)
KR (1) KR20050043996A (no)
AU (1) AU3447097A (no)
BR (1) BR9710030A (no)
CA (1) CA2258158A1 (no)
CZ (1) CZ438398A3 (no)
HU (1) HU224558B1 (no)
IL (1) IL127692A0 (no)
NO (1) NO986202L (no)
SK (1) SK181098A3 (no)
WO (1) WO1998000524A1 (no)

Families Citing this family (97)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2625279A1 (en) * 1995-08-30 1997-03-06 Genzyme Corporation Chromatographic purification of adenovirus and aav
US6485958B2 (en) * 1996-07-01 2002-11-26 Gencell Sa Method for producing recombinant adenovirus
US7732129B1 (en) 1998-12-01 2010-06-08 Crucell Holland B.V. Method for the production and purification of adenoviral vectors
JP4492826B2 (ja) * 1996-11-20 2010-06-30 イントロジェン セラピューティクス,インコーポレイテッド アデノウイルスベクターの産生および精製のための改良された方法
US20080261289A1 (en) * 1996-12-13 2008-10-23 Schering-Plough Corporation Compositions comprising viruses and methods for concentrating virus preparations
US6544769B1 (en) * 1996-12-13 2003-04-08 Schering Corporation Compostions comprising viruses and methods for concentrating virus preparations
AU5370598A (en) 1996-12-13 1998-07-03 Schering Corporation Methods for purifying viruses
US6146891A (en) 1997-01-31 2000-11-14 Schering Corporation Methods for cultivating cells and propagating viruses
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US6989264B2 (en) * 1997-09-05 2006-01-24 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
CA2328462C (en) 1998-04-22 2010-11-09 Genvec, Inc. Efficient purification of adenovirus
AU770672B2 (en) * 1998-05-27 2004-02-26 University Of Florida Method of preparing recombinant adeno-associated virus compositions by using an iodixananol gradient
WO1999061643A1 (en) * 1998-05-27 1999-12-02 University Of Florida Method of preparing recombinant adeno-associated virus compositions by using an iodixananol gradient
US6689600B1 (en) 1998-11-16 2004-02-10 Introgen Therapeutics, Inc. Formulation of adenovirus for gene therapy
IL143381A0 (en) * 1998-12-31 2002-04-21 Aventis Pharma Sa Method for separating viral particles
FR2788064B1 (fr) * 1998-12-31 2003-01-31 Aventis Pharma Sa Methode de separation de particules virales
EP1155120B1 (fr) 1999-02-22 2006-07-05 Transgene S.A. Procede d'obtention d'une preparation virale purifiee
US6783939B2 (en) * 2000-07-07 2004-08-31 Alphavax, Inc. Alphavirus vectors and virosomes with modified HIV genes for use in vaccines
ES2564553T3 (es) * 2001-08-08 2016-03-23 The Trustees Of The University Of Pennsylvania Procedimiento para purificar vectores víricos que tienen proteínas que se unen a ácido siálico
JP4790984B2 (ja) * 2001-09-06 2011-10-12 アルファヴァックス,インコーポレイテッド アルファウイルスレプリコンベクター系
WO2003049763A1 (en) 2001-12-12 2003-06-19 Fh Faulding & Co Limited Composition for the preservation of viruses
CN1617745A (zh) * 2002-01-18 2005-05-18 舍林股份公司 稳定的腺病毒配方
US20030224354A1 (en) * 2002-05-30 2003-12-04 Introgen Therapeutics Inc. Quantifying viral particles with intrinsic fluorescence
EP1371723A1 (en) * 2002-06-12 2003-12-17 Procorde GmbH Process for preparing an adenovirus-containing preparation
US20040106184A1 (en) * 2002-08-28 2004-06-03 Introgen Therapeutics Inc. Chromatographic methods for adenovirus purification
CA2509973C (en) 2002-12-13 2013-02-26 Alphavax, Inc. Multi-antigenic alphavirus replicon particles and methods
DK3246399T3 (da) * 2002-12-13 2021-10-04 Alphavax Inc Alfavirus-partikler og fremgangsmåder til fremstilling
MXPA05010007A (es) * 2003-03-20 2006-03-10 Alphavax Inc Replicones de alfavirus y construcciones auxiliares mejorados.
US20040229335A1 (en) 2003-05-15 2004-11-18 Introgen Therapeutics, Inc. Methods and compositions for the production of adenoviral vectors
CA2529053A1 (en) * 2003-06-18 2004-12-29 Onyx Pharmaceuticals, Inc. Method for purifying virus
WO2005007689A1 (en) * 2003-07-11 2005-01-27 Alphavax, Inc. Alphavirus-based cytomegalovirus vaccines
DE602004031681D1 (de) 2003-07-21 2011-04-14 Transgene Sa Multifunktionelle Cytokine
JP2007522814A (ja) * 2004-02-23 2007-08-16 クルセル ホランド ベー ヴェー ウイルスの精製方法
CA2567254C (en) 2004-05-18 2012-03-13 Alphavax, Inc. Tc-83-derived alphavirus vectors, particles and methods
WO2006052302A2 (en) * 2004-11-03 2006-05-18 Introgen Therapeutics Inc. Method of producing and purifying adenoviral vectors
US8900804B2 (en) * 2004-11-22 2014-12-02 Cedars-Sinai Medical Center Methods and solutions for tissue preservation
US7488848B2 (en) 2005-03-21 2009-02-10 Virobay, Inc. Alpha ketoamide compounds as cysteine protease inhibitors
ES2317517T5 (es) * 2005-04-11 2016-01-21 Crucell Holland B.V. Purificación de virus usando ultrafiltración
WO2008089448A2 (en) 2007-01-19 2008-07-24 Cornell Presearch Foundation, Inc. Methods and compositions for promoting survival & proliferation of endothelial cells & stimulating angiogenesis
AU2008209759B2 (en) 2007-01-30 2013-03-21 Transgene S.A. Papillomavirus E2 polypeptide used for vaccination
ES2670813T3 (es) 2007-06-21 2018-06-01 Alphavax, Inc. Partículas de replicón de alfavirus para uso en vacunación
MX2011004292A (es) * 2008-11-03 2011-05-31 Crucell Holland Bv Metodo para la produccion de vectores adenovirales.
CA2770075C (en) 2009-08-07 2021-08-24 Perrine Martin Composition for treating hbv infection
PL2488636T3 (pl) 2009-10-15 2014-07-31 Crucell Holland Bv Sposób oczyszczania cząsteczek adenowirusów z hodowli o wysokim zagęszczeniu komórkowym
AU2010305768B2 (en) 2009-10-15 2015-05-14 Crucell Holland B.V. Process for adenovirus purification from high cell density cultures
ES2578514T3 (es) 2010-02-15 2016-07-27 Crucell Holland B.V. Método para la producción de vectores adenovíricos
KR101549296B1 (ko) * 2010-04-14 2015-09-01 이엠디 밀리포어 코포레이션 고역가 고순도 바이러스 스톡의 생산 방법 및 이의 사용 방법
ES2627117T3 (es) 2010-12-09 2017-07-26 Institut Pasteur Método basado en MGMT para obtener un rendimiento elevado de expresión de proteínas recombinantes
TWI575070B (zh) 2011-07-12 2017-03-21 傳斯堅公司 Hbv聚合酶突變體
WO2013045658A1 (en) 2011-09-29 2013-04-04 Transgene Sa Immunotherapy composition and regimen for treating hepatitis c virus infection
WO2013045668A2 (en) 2011-09-29 2013-04-04 Transgene Sa Immunotherapy composition and regimen for treating hepatitis c virus infection
NZ628385A (en) 2012-03-12 2016-09-30 Crucell Holland Bv Batches of recombinant adenovirus with altered terminal ends
NZ630649A (en) 2012-03-22 2016-12-23 Janssen Vaccines & Prevention Bv Vaccine against rsv
US9125870B2 (en) 2012-03-22 2015-09-08 Crucell Holland B.V. Vaccine against RSV
US20150150963A1 (en) 2012-06-05 2015-06-04 The Australian National University Vaccination with interleukin-4 antagonists
WO2014066443A1 (en) 2012-10-23 2014-05-01 Emory University Gm-csf and il-4 conjugates, compositions, and methods related thereto
EP2961417A1 (en) * 2013-02-28 2016-01-06 PsiOxus Therapeutics Limited A process for the production of adenovirus
KR102089121B1 (ko) 2013-03-14 2020-03-13 더 솔크 인스티튜트 포 바이올로지칼 스터디즈 종양살상형 아데노바이러스 조성물
EP2988780B1 (en) 2013-04-25 2018-12-26 Janssen Vaccines & Prevention B.V. Stabilized soluble prefusion rsv f polypeptides
CN105408348B (zh) 2013-06-17 2021-07-06 扬森疫苗与预防公司 稳定化的可溶性融合前rsv f多肽
GB201415579D0 (en) * 2014-09-03 2014-10-15 Psioxus Therapeutics Ltd A process
AU2015279552B2 (en) * 2014-06-27 2021-01-21 Angiocrine Bioscience, Inc. Neural cells expressing adenovirus E4ORF1, and methods of making and using the same
WO2016030489A2 (en) * 2014-08-27 2016-03-03 Psioxus Therapeutics Limited A process for the production of adenovirus
WO2016114992A2 (en) 2015-01-13 2016-07-21 Alfa Wassermann, Inc. Methods of purifying adeno-associated virus (aav) and/or recombinant adeno-associated virus (raav) and gradients and flow-through buffers therefore
WO2016131945A1 (en) 2015-02-20 2016-08-25 Transgene Sa Combination product with autophagy modulator
EP3283634B1 (en) 2015-04-14 2019-05-22 Janssen Vaccines & Prevention B.V. Recombinant adenovirus expressing two transgenes with a bidirectional promoter
PL3319633T3 (pl) 2015-07-07 2021-04-19 Janssen Vaccines & Prevention B.V. Szczepionka przeciwko rsv
KR20180026734A (ko) 2015-07-07 2018-03-13 얀센 백신스 앤드 프리벤션 비.브이. 안정화된 가용성 융합-전 rsv f 폴리펩티드
EP3390645B1 (en) 2016-02-23 2022-09-14 Salk Institute for Biological Studies Exogenous gene expression in therapeutic adenovirus for minimal impact on viral kinetics
CA3013637A1 (en) * 2016-02-23 2017-08-31 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
WO2017174568A1 (en) 2016-04-05 2017-10-12 Janssen Vaccines & Prevention B.V. Stabilized soluble pre-fusion rsv f proteins
MX2018012095A (es) 2016-04-05 2019-01-10 Janssen Vaccines & Prevention Bv Vacuna contra vrs.
US20190134190A1 (en) 2016-05-04 2019-05-09 Transgene Sa Combination therapy with cpg tlr9 ligand
JP7046835B2 (ja) 2016-05-12 2022-04-04 ヤンセン ファッシンズ アンド プリベンション ベーフェー 強力でバランスのとれた双方向性プロモーター
DK3464331T3 (da) 2016-05-30 2021-01-18 Janssen Vaccines & Prevention Bv Stabiliserede præfusions-rsv f-proteiner
BR112018075969A2 (pt) 2016-06-20 2019-04-02 Janssen Vaccines & Prevention B.V. promotor bidirecional potente e equilibrado
JP7229151B2 (ja) 2016-07-14 2023-02-27 ヤンセン ファッシンズ アンド プリベンション ベーフェー Hpvワクチン
WO2018069316A2 (en) 2016-10-10 2018-04-19 Transgene Sa Immunotherapeutic product and mdsc modulator combination therapy
CN110418650A (zh) 2016-11-16 2019-11-05 免疫治疗有限公司 用于治疗过敏的核酸
EP3532082A4 (en) 2016-12-12 2020-08-26 Salk Institute for Biological Studies SYNTHETIC ADENOVIRUS TUMOR TARGETING AND THEIR USES
US11034978B2 (en) 2017-02-09 2021-06-15 Janssen Vaccines & Prevention B.V. Potent and short promoter for expression of heterologous genes
WO2018195527A1 (en) 2017-04-22 2018-10-25 Immunomic Therapeutics, Inc. Improved lamp constructs
WO2018204534A1 (en) 2017-05-02 2018-11-08 Immunomic Therapeutics, Inc. Lamp (lysosomal associated membrane protein) constructs comprising cancer antigens
JP2020519663A (ja) 2017-05-17 2020-07-02 ヤンセン ファッシンズ アンド プリベンション ベーフェーJanssen Vaccines & Prevention B.V. Rsv感染に対する防御免疫を誘導するための方法及び組成物
EP3681533A1 (en) 2017-09-15 2020-07-22 Janssen Vaccines & Prevention B.V. Method for the safe induction of immunity against rsv
WO2019222281A1 (en) 2018-05-15 2019-11-21 Immunomic Therapeutics, Inc Improved lamp constructs comprising allergens
TW202043256A (zh) 2019-01-10 2020-12-01 美商健生生物科技公司 前列腺新抗原及其用途
JP2022521268A (ja) 2019-02-21 2022-04-06 アンリーシュ イミュノ オンカリティクス,インコーポレイテッド 腫瘍溶解性アデノウイルスベクター及び使用法
JP2022551732A (ja) 2019-10-18 2022-12-13 イミュノミック セラピューティックス, インコーポレイテッド がん抗原を含む改良lamp構築物
CN114980920A (zh) 2019-11-18 2022-08-30 詹森生物科技公司 基于突变体calr和jak2的疫苗及其用途
TW202144388A (zh) 2020-02-14 2021-12-01 美商健生生物科技公司 在卵巢癌中表現之新抗原及其用途
TW202144389A (zh) 2020-02-14 2021-12-01 美商健生生物科技公司 在多發性骨髓瘤中表現之新抗原及其用途
US20210315986A1 (en) 2020-04-13 2021-10-14 Janssen Biotech, Inc. Psma and steap1 vaccines and their uses
WO2022009049A1 (en) 2020-07-06 2022-01-13 Janssen Biotech, Inc. Prostate neoantigens and their uses
US20230029453A1 (en) 2020-07-06 2023-02-02 Janssen Biotech, Inc. Prostate Neoantigens And Their Uses
EP4176087A1 (en) 2020-07-06 2023-05-10 Janssen Biotech, Inc. A method for determining responsiveness to prostate cancer treatment
WO2023201201A1 (en) 2022-04-10 2023-10-19 Immunomic Therapeutics, Inc. Bicistronic lamp constructs comprising immune response enhancing genes and methods of use thereof

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5173418A (en) * 1985-05-10 1992-12-22 Benzon Pharma, A/S Production in Escherichia coli of extracellular Serratia spp. hydrolases
FR2705686B1 (fr) * 1993-05-28 1995-08-18 Transgene Sa Nouveaux adénovirus défectifs et lignées de complémentation correspondantes.
WO1995019427A1 (en) 1994-01-12 1995-07-20 Genetic Therapy, Inc. Purification of retroviral vectors
FR2716893B1 (fr) 1994-03-03 1996-04-12 Rhone Poulenc Rorer Sa Virus recombinants, leur préparation et leur utilisation thérapeutique.
WO1995025789A1 (en) * 1994-03-22 1995-09-28 The Immune Response Corporation Highly efficient production and isolation of viral particles
US5837520A (en) * 1995-03-07 1998-11-17 Canji, Inc. Method of purification of viral vectors
US6485958B2 (en) * 1996-07-01 2002-11-26 Gencell Sa Method for producing recombinant adenovirus
JP4492826B2 (ja) 1996-11-20 2010-06-30 イントロジェン セラピューティクス,インコーポレイテッド アデノウイルスベクターの産生および精製のための改良された方法
US6261823B1 (en) * 1996-12-13 2001-07-17 Schering Corporation Methods for purifying viruses
IL143381A0 (en) * 1998-12-31 2002-04-21 Aventis Pharma Sa Method for separating viral particles

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9800524A1 *

Also Published As

Publication number Publication date
CZ438398A3 (cs) 1999-03-17
HUP0001271A1 (hu) 2000-08-28
US6485958B2 (en) 2002-11-26
AU3447097A (en) 1998-01-21
SK181098A3 (en) 1999-07-12
BR9710030A (pt) 1999-08-10
JP2000514290A (ja) 2000-10-31
NO986202L (no) 1999-02-15
HU224558B1 (hu) 2005-10-28
US20050287657A1 (en) 2005-12-29
KR20050043996A (ko) 2005-05-11
NO986202D0 (no) 1998-12-30
US20030143730A1 (en) 2003-07-31
US20020028497A1 (en) 2002-03-07
CA2258158A1 (fr) 1998-01-08
WO1998000524A1 (fr) 1998-01-08
US6905862B2 (en) 2005-06-14
IL127692A0 (en) 1999-10-28
HUP0001271A3 (en) 2002-01-28

Similar Documents

Publication Publication Date Title
EP0944717A1 (fr) Procede de production d'adenovirus recombinants
WO1998000524A9 (fr) Procede de production d'adenovirus recombinants
CA2230655C (en) Chromatographic purification of adenovirus and aav
KR100762033B1 (ko) 바이러스 입자의 분리방법
Sviben et al. Recovery of infective virus particles in ion-exchange and hydrophobic interaction monolith chromatography is influenced by particle charge and total-to-infective particle ratio
CA2181602A1 (fr) Procede de preparation de virus adeno-associes (aav) recombinants et utilisations
Segura et al. Chromatography purification of canine adenoviral vectors
US20050260168A1 (en) Compositions comprising viruses and methods for concentrating virus preparations
FR2707664A1 (fr) Vecteurs viraux et utilisation en thérapie génique.
JP2007117003A (ja) ウイルス中空粒子の迅速除去および精製方法
FR2750433A1 (fr) Procede de production d'adenovirus recombinants
AU778287B2 (en) Method for producing recombinant adenovirus
AU2004201075B2 (en) Method for producing recombinant adenovirus
Dietl et al. An efficient capture strategy for the purification of human adenovirus type 5 from cell lysates
KR100514589B1 (ko) 재조합아데노바이러스의제조방법
MXPA99000230A (en) Procedure of production of adenovirus recombinan
FR2788064A1 (fr) Methode de separation de particules virales
FR2729674A1 (fr) Cellules pour la production d'adenovirus recombinants

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19990104

AK Designated contracting states

Kind code of ref document: A1

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

AX Request for extension of the european patent

Free format text: SI PAYMENT 19990104

RAP3 Party data changed (applicant data changed or rights of an application transferred)

Owner name: AVENTIS PHARMA S.A.

17Q First examination report despatched

Effective date: 20031112

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: GENCELL S.A.

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CENTELION

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

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20070627