US20040229335A1 - Methods and compositions for the production of adenoviral vectors - Google Patents

Methods and compositions for the production of adenoviral vectors Download PDF

Info

Publication number
US20040229335A1
US20040229335A1 US10/439,278 US43927803A US2004229335A1 US 20040229335 A1 US20040229335 A1 US 20040229335A1 US 43927803 A US43927803 A US 43927803A US 2004229335 A1 US2004229335 A1 US 2004229335A1
Authority
US
United States
Prior art keywords
adenovirus
media
host cells
medium
cells
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.)
Abandoned
Application number
US10/439,278
Other languages
English (en)
Inventor
Shuyuan Zhang
Hai Pham
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.)
Janssen Vaccines and Prevention BV
Original Assignee
Introgen Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Introgen Therapeutics Inc filed Critical Introgen Therapeutics Inc
Priority to US10/439,278 priority Critical patent/US20040229335A1/en
Assigned to INTROGEN THERAPEUTICS, INC. reassignment INTROGEN THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PHAM, HAI, ZHANG, SHUYUAN
Priority to JP2006533032A priority patent/JP2007500015A/ja
Priority to CNA2004800185873A priority patent/CN1816620A/zh
Priority to CA002525603A priority patent/CA2525603A1/en
Priority to PCT/US2004/015009 priority patent/WO2004104190A2/en
Priority to EP04752113A priority patent/EP1627057A2/en
Priority to AU2004241421A priority patent/AU2004241421A1/en
Priority to KR1020057021606A priority patent/KR20060028388A/ko
Publication of US20040229335A1 publication Critical patent/US20040229335A1/en
Priority to US11/079,986 priority patent/US7419808B2/en
Priority to IL171904A priority patent/IL171904A0/en
Assigned to CRUCELL HOLLAND B.V. reassignment CRUCELL HOLLAND B.V. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: INTROGEN THERAPEUTICS, INC.
Abandoned legal-status Critical Current

Links

Images

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • 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

Definitions

  • the present invention relates generally to the fields of cell culture and virus production. More particularly, it concerns improved methods for the culturing of mammalian cells, infection of those cells with adenovirus and the production and purification of infectious adenovirus particles therefrom.
  • Viruses are highly efficient at nucleic acid delivery to specific cell types, while often avoiding detection by the infected host's immune system. These features make certain viruses attractive candidates as gene-delivery vehicles for use in gene therapies (Robbins and Ghivizzani, 1998; Cristiano et al., 1998). Modified adenoviruses that are replication incompetent and therefore non-pathogenic are being used as vehicles to deliver therapeutic genes for a number of metabolic and oncologic disorders. These adenoviral vectors may be particularly suitable for disorders such as cancer that would best be treated by transient therapeutic gene expression since the DNA is not integrated into the host genome and the transgene expression is limited. Adenoviral vector may also be of significant benefit in gene replacement therapies, wherein a genetic or metabolic defect or deficiency is remedied by providing for expression of a replacement gene encoding a product that remedies the defect or deficiency.
  • Adenoviruses can be modified to efficiently deliver a therapeutic or reporter transgene to a variety of cell types.
  • Recombinant adenoviruses types 2 and 5 Ad2 and AdV5, respectively
  • Ad2 and AdV5 are among those currently being developed for gene therapy.
  • Both Ad2 and AdV5 belong to a subclass of adenovirus that are not associated with human malignancies.
  • the hybrid adenoviral vector AdV5/F35 has been developed and proven of great interest in gene therapies and related studies (Yotnda et al., 2001).
  • Recombinant adenoviruses are capable of providing extremely high levels of transgene delivery.
  • the efficacy of this system in delivering a therapeutic transgene in vivo that complements a genetic imbalance has been demonstrated in animal models of various disorders (Watanabe, 1986; Tanzawa et al., 1980; Golasten et al., 1983; Ishibashi et al., 1993; and S. Ishibashi et al., 1994).
  • CFTR cystic fibrosis transmembrane regulator
  • CFTR cystic fibrosis transmembrane regulator
  • adenoviruses are produced in commercially available tissue culture flasks or “cellfactories.”
  • Adenoviral vector production has generally been performed in culture devices that supply culture surfaces for attachment of the HEK293 cells, such as T-flasks.
  • Virus infected cells are harvested and freeze-thawed to release the viruses from the cells in the form of crude cell lysate.
  • the produced crude cell lysate (CCL) is then purified by double CsCl gradient ultracentrifugation.
  • the typically reported virus yield from 100 single tray cell factories is about 6 ⁇ 10 12 PFU.
  • New scaleable and validatable production and purification processes have to be developed to meet the increasing demand.
  • HEK293 cells which complement the adenoviral vector E1 deletion in trans. Because of the anchorage dependency of the HEK293 cells, adenoviral vector production has generally been performed in culture devices that supply culture surfaces for attachment of the HEK293 cells, such as T-flasks, multilayer CellfactoriesTM, and the large scale CellCubeTM bioreactor system. Recently, the HEK293 cells have been adapted to suspension culture in a variety of serum free media allowing production of adenoviral vectors in suspension bioreactors. Complete medium exchange at the time of virus infection using centrifugation is difficult to perform on a large scale. In addition, the shear stress associated with medium recirculation required for external filtration devices is likely to have a detrimental effect on host cells in a protein-free medium.
  • Improved methods for adenoviral vector production can include improved techniques to make production more efficient, or optimization of operating conditions to increase adenoviral vector production.
  • the present invention is based on the discovery that when adenovirus is produced by infecting host cells grown in media, adenovirus production is maximized at infection temperatures slightly lower than 37° C.
  • the identification of the optimal infection temperature range for adenovirus infection represents a significant improvement in the technology for producing adenoviral vectors for gene therapy.
  • the present invention is based on the discovery that when host cells are grown in a bioreactor, the dilution step wherein fresh media is exchanged can be combined with the adenovirus infection step. More specifically, there is no need for a separate medium exchange step. Thus, the technique for mass production of adenovirus can be more efficiently performed by combining two steps into a single step. Approximately equivalent virus productivity was attained with this method of dilution/infection relative to the standard method of centrifugation and complete medium exchange prior to infection.
  • the present invention is designed to take advantage of these newly discovered improvements by providing for improved techniques related to adenovirus production and purification.
  • the invention relates to methods related to the production and purification of adenovirus, and compositions of adenovirus that are produced and purified in accordance with these new discoveries.
  • Methods of the invention includes methods for producing an adenovirus, including: (1) preparing an adenovirus preparation, including the steps of growing host cells in media; and infecting the host cells with an adenovirus at a growth-permissive temperature of less than 37° C.; and (2) isolating adenovirus from the adenovirus preparation.
  • infecting the host cells with an adenovirus is conducted at a temperature greater than 31° C. but less than 37° C.
  • infecting the host cells with an adenovirus may be conducted at a temperature within the range of 32° C. to 36° C., 33° C. to 36° C., 34° C. to 36° C., 35° C.
  • infecting the host cells with an adenovirus is conducted at a temperature within the range of 32° C. to less than 37° C., 33° C. to less than 37° C., 34° C. to less than 37° C., 35° C. to less than 37° C., 36° C. to less than 37° C., or any range of temperature or increments of temperature derivable therein.
  • infecting the host cells with an adenovirus may be conducted at a temperature of about 36° C., about 35° C., about 34° C., about 33° C., about 32° C., or any range of temperature or increments of temperature derivable therein.
  • any media for growing host cells is contemplated, as long as the media is capable of supporting the growth of host cells.
  • the media is DMEM+2% FBS.
  • the present invention also contemplates that other substances may be added to the media to support host cell growth.
  • One of skill in the art would be familiar with the range of substances and additives that may be used to promote cell growth.
  • the glucose concentration in said media is maintained between about 0.5 and about 3.0 gm glucose/liter.
  • inventions of the present invention pertain to methods for producing an adenovirus, including: (1) preparing an adenovirus preparation, including the steps of growing host cells in media in a bioreactor and initiating virus infection by diluting the host cells with fresh media and adenovirus; and (2) isolating adenovirus from the adenovirus preparation.
  • Any bioreactor known to those of skill in the art that is capable of supporting host cell growth is contemplated for use in the present invention. A detailed discussion of various types of bioreactors is presented below in other parts of the specification.
  • the various embodiments of the present invention requiring a bioreactor contemplate that any type of media may be used in conjunction with the bioreactor, as long as the media is capable of supporting cell growth in the bioreactor.
  • the media may be serum-free media.
  • the media is protein-free media.
  • the media is CD293 media.
  • the host cells may be grown in an anchorage-dependent culture or a non-anchorage-dependent (suspension) culture.
  • the bioreactor comprises a bioreactor that uses axial rocking of a planar platform to induce wave motions inside of the bioreactor.
  • wave motions are induced inside of a sterilized polyethylene bag wherein the host cells are located.
  • the bioreactor is a disposable bioreactor. Any size of bioreactor is contemplated by the present invention.
  • the bioreactor may be a 10 L bioreactor.
  • the bioreactor may be a commercially-available bioreactor.
  • the bioreactor may be a Wave Bioreactor® (Wave Biotech, LLC, Bedminster, N.J.).
  • the operating conditions of the cell culture may be monitored or measured by any technique known to those of skill in the art. Examples of such conditions which may be monitored include pH of the media and dissolved oxygen tension of the media.
  • Some embodiments of the present invention pertaining to methods of producing an adenovirus also involve processing and treating the media by any method known to those of skill in the art.
  • the methods for producing an adenovirus involve perfusing the media through a filter.
  • the filter may be a filter that is internal to the bioreactor system, or the filter may be incorporated so that it is external to the bioreactor.
  • the filter is a floating flat filter.
  • the floating flat filter may be used to remove spent media from the bioreactor. Any method known to those of skill in the art may be used to monitor and maintain media volume.
  • culture volume is maintained by a load cell used to trigger fresh media addition.
  • media may or may not be perfused into the culture of host cells.
  • media is perfused beginning on day 3 of host cell growth.
  • One of skill in the art would be familiar with the wide range of techniques and apparatus available for perfusing media into a cell culture system.
  • the step of diluting host cells with fresh media may be combined with the adenovirus infection step. This is based on the inventors' discovery that these two steps can be efficiently combined to provide for excellent yields of adenoviral vectors.
  • the invention contemplates use of any method of dilution known to those of skill in the art.
  • the host cells are diluted 2-fold to 50-fold with fresh media and adenovirus. In other embodiments, the host cells are diluted 10-fold with fresh media and adenovirus.
  • the initiating of virus infection of the host cells may be accomplished by any method known to those of skill in the art.
  • the virus infection may take place in a second bioreactor.
  • virus infection of host cells may be accomplished by adding 20-100 vp/host cell.
  • virus infection involves adding about 50 vp/host cell.
  • Virus infection may be allowed to proceed for any duration of time.
  • One of skill in the art would be familiar with techniques pertaining to monitoring the progress of virus infection.
  • virus infection is allowed to proceed for about 4 days.
  • the isolating of the adenovirus from the adenovirus preparation occurs at about 4 days after viral infection is completed.
  • host cells In the embodiments of the present invention that involve production of adenovirus, use of host cells is contemplated. Any cell type can be used as a host cell, as long as the cell is capable of supporting replication of adenovirus.
  • the host cells complement the growth of the replication-deficient adenovirus.
  • the replication-deficient adenovirus may be an adenovirus that lacks at least a portion of the E1-region, or it may be an adenovirus that lacks at least a portion of the E1A and/or E1B region.
  • the host cells for example, may be 293, HEK293, PER.C6, 911, and IT293SF cells. In certain embodiments of the present invention, the host cells are HEK293 cells.
  • the adenovirus is a recombinant adenovirus.
  • the recombinant adenovirus may encode a recombinant gene that is operatively linked to a promoter.
  • Any promoter known to those of skill in the art can be used, as long as the promoter is capable of functioning as a promoter.
  • the promoter is an SV40 E1, RSV LTR, ⁇ -actin, CMV-IE, adenovirus major late, polyoma F9-1, or tyrosinase promoter.
  • the adenovirus is an adenovirus encoding a recombinant gene
  • any recombinant gene particularly a therapeutic gene
  • the recombinant gene may be selected from the group consisting of antisense ras, antisense myc, antisense raf, antisense erb, antisense src, antisense fms, antisense jun, antisense trk, antisense ret, antisense gsp, antisense hst, antisense bcl antisense abl, Rb, CFTR, p16, p21, p27, p57, p73, C-CAM, APC, CTS-1, zac1, scFV ras, DCC, NF-1, NF-2, WT-1, MEN-I, MEN-II, BRCA1,
  • the recombinant gene is antisense ras, antisense myc, antisense raf, antisense erb, antisense src, antisense fms, antisense jun, antisense trk, antisense ret, antisense gsp, antisense hst, antisense bcl antisense abl, Rb, CFTR, p16, p21, p27, p57, p73, C-CAM, APC, CTS-1, zac1, scFV ras, DCC, NF-1, NF-2, WT-1, MEN-I, MEN-II, BRCA1, VHL, MMAC1, FCC, MCC, BRCA2, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11
  • the recombinant gene is a gene encoding an ACP desaturase, an ACP hydroxylase, an ADP-glucose pyrophorylase, an ATPase, an alcohol dehydrogenase, an amylase, an amyloglucosidase, a catalase, a cellulase, a cyclooxygenase, a decarboxylase, a dextrinase, an esterase, a DNA polymerase, an RNA polymerase, a hyaluron synthase, a galactosidase, a glucanase, a glucose oxidase, a GTPase, a helicase, a hemicellulase, a hyaluronidase, an integrase, an invertase, an isomerase, a kinase,
  • the recombinant gene is a gene encoding carbamoyl synthetase I, ornithine transcarbamylase, arginosuccinate synthetase, arginosuccinate lyase, arginase, fumarylacetoacetate hydrolase, phenylalanine hydroxylase, alpha-1 antitrypsin, glucose-6-phosphatase, low-density-lipoprotein receptor, porphobilinogen deaminase, factor VIII, factor IX, cystathione beta.-synthase, branched chain ketoacid decarboxylase, albumin, isovaleryl-CoA dehydrogenase, propionyl CoA carboxylase, methyl malonyl CoA mutase, glutaryl CoA dehydrogenase, insulin, beta.-glucosidase, pyruvate carboxylase, hepatic phospho
  • the recombinant gene may encode growth hormone, prolactin, placental lactogen, luteinizing hormone, follicle-stimulating hormone, chorionic gonadotropin, thyroid-stimulating hormone, leptin, adrenocorticotropin, angiotensin I, angiotensin II, ⁇ -endorphin, ⁇ -melanocyte stimulating hormone, cholecystokinin, endothelin I, galanin, gastric inhibitory peptide, glucagon, insulin, lipotropins, neurophysins, somatostatin, calcitonin, calcitonin gene related peptide, ⁇ -calcitonin gene related peptide, hypercalcemia of malignancy factor, parathyroid hormone-related protein, parathyroid hormone-related protein, glucagon-like peptide, pancreastatin, pancreatic peptide, peptide YY, PHM, secretin, vasoactive intestinal peptid
  • Certain of the embodiments of the present invention pertain to methods of producing an adenovirus that involve isolating the adenovirus from an adenovirus preparation. Any method of isolating the adenovirus from the adenovirus preparation known to those of skill in the art is contemplated by the present invention.
  • the host cells are harvested following infection but prior to lysis by the adenovirus, and lysing the host cells is performed by freeze-thaw, autolysis, or detergent lysis.
  • the methods of producing adenovirus involve reducing the concentration of contaminating nucleic acids in the adenovirus preparation.
  • the adenovirus that is isolated is placed into a pharmaceutically acceptable composition.
  • a pharmaceutically acceptable composition One of skill in the art would be familiar with the extensive methods and techniques employed in preparing pharmaceutically acceptable compositions. Any pharmaceutical composition into which adenovirus can be formulated is contemplated by the present invention.
  • certain embodiments of the invention pertain to pharmaceutical preparation of adenovirus for oral administration, topical administration, or intravenous administration.
  • Certain embodiments of the present invention involve methods of purifying an adenovirus that has been isolated from an adenovirus preparation.
  • purifying the adenovirus may involve a chromatography step.
  • the chromatography step include subjecting the adenovirus to more than one chromatographic separation whereas in other embodiments the chromatography step involves subjecting the adenovirus to only one chromatographic separation.
  • the chromatographic separation includes ion exchange chromatography.
  • virus production may be analyzed using HPLC. Any technique for analyzing virus production known to those of skill is contemplated by the present invention.
  • the methods for producing an adenovirus disclosed above and elsewhere in this specification concern methods for isolating and purifying an adenovirus that involve obtaining a purified adenovirus composition having one or more of the following properties: (1) a virus titer of between 1 ⁇ 10 9 and about 1 ⁇ 10 13 pfu/ml; (2) a virus particle concentration between about 1 ⁇ 10 10 and about 2 ⁇ 10 13 particles/ml; (3) a particle:pfu ration between about 10 and about 60; (4) having less than 50 ng BSA per 1 ⁇ 10e12 viral particles; (5) between about 50 pg and 1 ng of contaminating human DNA per 1 ⁇ 10 12 viral particles; (6) a single HPLC elution peak consisting essentially of 97% to 99% of the area under the peak.
  • the adenovirus composition prepared in accordance with the steps discussed above includes between 5 ⁇ 10 14 and 1 ⁇ 10 18 viral particles.
  • the composition is a virus titer of between 1 ⁇ 10 9 and about 1 ⁇ 10 13
  • the methods of producing an adenovirus discussed above and elsewhere in this specification involve isolating the adenovirus from the adenovirus preparation comprises the steps of: (1) subjecting the adenovirus preparation to chromatography on a first chromatographic medium, whereby adenovirus particles are retained on the first chromatographic medium; (2) eluting adenovirus particles from the first chromatographic medium to produce an eluate of adenovirus particles; (3) subjecting adenovirus particles from the eluate to chromatography on a second chromatographic medium, wherein the second chromatographic medium retains one or more contaminants from the eluate and wherein the second chromotographic medium is not solely a size exclusion medium; and (4) collecting adenovirus particles from the eluate.
  • the first chromatographic medium is selected from the group consisting of an anion exchange medium, cation exchange medium, immobilized metal affinity medium, sulfated affinity media, immunoaffinity medium, heparin affinity medium, hydroxyapetite medium, and hydrophobin interaction medium.
  • the second chromatographic medium is selected from the group consisting of cation exchange media, anion exchange media, immobilized metal affinity media, sulfated affinity media, dye affinity media, hydroxyapetite media, immunoaffinity media, heparin affinity media, and hydrophobic interaction media.
  • the methods of producing an adenovirus discussed above and elsewhere in this specification involve isolating the adenovirus from the adenovirus preparation comprises the steps of: (1) subjecting the adenovirus preparation to chromatography on a first chromatographic medium, whereby contaminants from the adenovirus preparation are retained on the first chromatographic medium; (2) subjecting adenovirus particles remaining in the eluant to chromatography on a second chromatographic medium wheregy adenovirus particles from the eluant are retained on the second chromatographic medium, wherein when the second chromatographic medium is an anion exchange medium, then the first chromatographic medium is a medium other than a sulfonated polysaccharide affinity medium, and (3) eluting adenovirus particles from the second chromatographic medium.
  • the first chromatographic medium is selected from the group consisting of an anion exchange medium, cation exchange medium, immobilized metal affinity medium, sulfated affinity media, immunoaffinity medium, heparin affinity medium, hydroxyapetite medium, and hydrophobin interaction medium.
  • the second chromatographic medium is selected from the group consisting of cation exchange media, anion exchange media, immobilized metal affinity media, sulfated affinity media, dye affinity media, hydroxyapetite media, immunoaffinity media, heparin affinity media, and hydrophobic interaction media. Any chromatographic media known to those of skill in the art is contemplated by the present invention. One of skill in the art would be familiar with the range of media available to practice the claimed invention.
  • FIG. 1A show results of a study demonstrating the effect of temperature on Adp53 production (vp/flask).
  • FIG. 1B shows results of a study demonstrating the effect of temperature on Admda7 production (vp/flask).
  • FIG. Cell growth and viability in the bioreactor.
  • FIG. 3 Glucose and lactate concentrations (g/L) in media vs. days in culture.
  • FIG. 4 Diagram of a perfusion bioreactor system.
  • FIG. 5 Cell growth and viability in perfusion culture vs. days in culture.
  • FIG. 6 Glucose and lactate concentrations (g/L) in perfusion culture vs. days in culture.
  • adenoviral vectors can successfully be used in eukaryotic gene expression and vaccine development.
  • adenovirus can be used for gene therapy.
  • Successful studies in administering recombinant adenovirus to different tissues have proven the effectiveness of adenoviral vectors in gene therapy. This success has led to the use of such vectors in human clinical trials.
  • the present invention provides methods for the production and purification of large amounts of adenovirus for use in such therapies.
  • the inventors have made the surprising discovery that adenovirus production is maximized at temperatures slightly lower than 37° C.
  • the identification of the optimal infection temperature range for adenovirus infection represents a significant improvement in the technology for producing adenoviral vectors for gene therapy.
  • the inventors have discovered that when host cells are grown in a bioreactor, the dilution step wherein fresh media is exchanged can be combined with the adenovirus infection step without loss of virus production. More specifically, there is no need for a separate medium exchange step. Thus, the technique for mass production of adenovirus can be more efficiently performed by combining two steps into a single step.
  • the present invention is designed to take advantage of these improvements in large scale culturing systems and purification for the purpose of producing and purifying adenoviral vectors.
  • Methods for producing and purifying adenovirus using the novel findings of the inventors are set forth in detail below.
  • Adenoviruses comprise linear double stranded DNA, with a genome ranging from 30 to 35 kb in size (Reddy et al., 1998; Morrison et al, 1997; Chillon et al., 1999). There are over 50 serotypes of human adenovirus, and over 80 related forms which are divided into six families based on immunological, molecular, and functional criteria (Wadell et al, 1980). Physically, adenovirus is a medium-sized icosahedral virus containing a double-stranded, linear DNA genome which, for adenovirus type 5, is 35,935 base pairs (Chroboczek et al., 1992). Adenoviruses require entry into the host cell and transport of the viral genome to the nucleus for infection of the cell and replication of the virus.
  • Salient features of the adenovirus genome are an early region (E1, E2, E3 and E4 genes), an intermediate region (pIX gene, Iva2 gene), a late region (L1, L2, L3, L4 and L5 genes), a major late promoter (MLP), inverted-terminal-repeats (ITRs) and a ⁇ sequence (Zheng, et al., 1999; Robbins et al., 1998; Graham and Prevec, 1995).
  • the early genes E1, E2, E3 and E4 are expressed from the virus after infection and encode polypeptides that regulate viral gene expression, cellular gene expression, viral replication, and inhibition of cellular apoptosis.
  • the MLP is activated, resulting in the expression of the late (L) genes, encoding polypeptides required for adenovirus encapsidation.
  • the intermediate region encodes components of the adenoviral capsid.
  • Adenoviral inverted terminal repeats ITRs; 100-200 bp in length
  • ITRs are cis elements, function as origins of replication and are necessary for viral DNA replication.
  • the ⁇ sequence is required for the packaging of the adenoviral genome.
  • CAR Coxsackie Adenoviral Receptor
  • the endogenous cellular function of CAR has not yet been elucidated. Interaction between the fiber knob and CAR is sufficient for binding of the adenovirus to the cell surface.
  • subsequent interactions between the penton base and additional cell surface proteins, members of the ⁇ v integrin family, are necessary for efficient viral internalization. Disassembly of the adenovirus begins during internalization; the fiber proteins remain on the cell surface bound to CAR.
  • the remainder of the adenovirus is dissembled in a stepwise manner as the viral particle is transported through the cytoplasm to a pore complex at the nuclear membrane.
  • the viral DNA is extruded through the nuclear membrane into the nucleus where viral DNA is replicated, viral proteins are expressed, and new viral particles are assembled. Specific steps in this mechanism of adenoviral infection may be potential targets to modulate viral infection and gene expression.
  • the adenovirus used in the methods for producing an adenovirus may be a replication-deficient adenovirus.
  • the adenovirus may be a replication-deficient adenovirus lacking at least a portion of the E1 region.
  • the adenovirus may be lacking at least a portion of the E1A and/or E1B region.
  • the adenovirus is a recombinant adenovirus (discussed further below).
  • a “host cell” is defined as a cell that is capable of supporting replication of adenovirus. Any cell type for use as a host cell is contemplated by the present invention invention, as long as the cell is capable of supporting replication of adenovirus.
  • the host cells may be HEK293, PER.C6, 911, or IT293SF cells.
  • One of skill in the art would be familiar with the wide range of host cells that are available for use in methods for producing an adenovirus.
  • the generation and propagation of the adenoviral vectors depend on a unique helper cell line, designated 293, which was transformed from human embryonic kidney cells by Adenovirus serotype 5 (Ad5) DNA fragments and constitutively expresses E1 proteins (Graham et al., 1977). Since the E3 region is dispensable from the Ad genome (Jones and Shenk, 1978), the current Ad vectors, with the help of 293 cells, carry foreign DNA in either the E1, the E3 or both regions (Graham and Prevec, 1991; Bett et al., 1994).
  • Ad5 Adenovirus serotype 5
  • the host cells used in the various embodiments of the present invention may be derived, for example, from mammalian cells such as human embryonic kidney cells or primate cells.
  • mammalian cells such as human embryonic kidney cells or primate cells.
  • Other cell types might include, but are not limited to Vero cells, CHO cells or any eukaryotic cells for which tissue culture techniques are established as long as the cells are adenovirus permissive.
  • adenovirus permissive means that the adenovirus or adenoviral vector is able to complete the entire intracellular virus life cycle within the cellular environment.
  • the host cell may be derived from an existing cell line, e.g., from a 293 cell line, or developed de novo. Such host cells express the adenoviral genes necessary to complement in trans deletions in an adenoviral genome or which supports replication of an otherwise defective adenoviral vector, such as the E1, E2, E4, E5 and late functions. A particular portion of the adenovirus genome, the E1 region, has already been used to generate complementing cell lines. Whether integrated or episomal, portions of the adenovirus genome lacking a viral origin of replication, when introduced into a cell line, will not replicate even when the cell is superinfected with wild-type adenovirus.
  • a cell line according to the present invention will express E1 and/or E4.
  • Recombinant host cells which are host cells that express part of the adenoviral genome, are also contemplated for use as host cells in the present invention.
  • the term “recombinant” cell is intended to refer to a cell into which a gene, such as a gene from the adenoviral genome or from another cell, has been introduced. Therefore, recombinant cells are distinguishable from naturally-occurring cells which do not contain a recombinantly-introduced gene. Recombinant cells are thus cells having a gene or genes introduced through “the hand of man.”
  • Recombinant host cells lines are capable of supporting replication of adenovirus recombinant vectors and helper viruses having defects in certain adenoviral genes, i.e., are “permissive” for growth of these viruses and vectors.
  • the recombinant cell also is referred to as a helper cell because of the ability to complement defects in, and support replication of, replication-incompetent adenoviral vectors.
  • Examples of other useful mammalian cell lines that may be used with a replication competent virus or converted into complementing host cells for use with replication deficient virus are Vero and HeLa cells and cell lines of Chinese hamster ovary, W138, BHK, COS-7, HepG2, 3T3, RIN and MDCK cells.
  • selection systems that preclude growth of undesirable cells. This may be accomplished by virtue of permanently transforming a cell line with a selectable marker or by transducing or infecting a cell line with a viral vector that encodes a selectable marker. In either situation, culture of the transformed/transduced cell with an appropriate drug or selective compound will result in the enhancement, in the cell population, of those cells carrying the marker.
  • markers include, but are not limited to, HSV thymidine kinase, hypoxanthine-guanine phosphoribosyltransferase and adenine phosphoribosyltransferase genes, in tk-, hgprt- or aprt-cells, respectively.
  • anti-metabolite resistance can be used as the basis of selection for dhfr, that confers resistance to methotrexate; gpt, that confers resistance to mycophenolic acid; neo, that confers resistance to the aminoglycoside G418; and hygro, that confers resistance to hygromycin.
  • the second method that has been used is a gradual adaptation of 293A cells into suspension growth (Cold Spring Harbor Laboratories, 293S cells).
  • Garnier et al (1994) reported the use of 293S cells for production of recombinant proteins from adenoviral vectors. The authors found that 293S cells were much less clumpy in calcium-free media and a fresh medium exchange at the time of virus infection could significantly increase the protein production. It was found that glucose was the limiting factor in culture without medium exchange.
  • Certain embodiments of the present invention pertain to methods for producing an adenovirus that require the use of a bioreactor.
  • a “bioreactor” refers to any apparatus that can be used for the purpose of culturing cells. Growing cells according to the present invention in a bioreactor allows for large scale production of fully biologically-active cells capable of being infected by the adenoviral vectors of the present invention.
  • Bioreactors have been widely used for the production of biological products from both suspension and anchorage dependent cell cultures.
  • the most widely used producer cells for adenoviral vector production are anchorage dependent human embryonic kidney cells (293 cells).
  • Bioreactors to be developed for adenoviral vector production should have the characteristic of high volume-specific culture surface area in order to achieve high producer cell density and high virus yield.
  • Microcarrier cell culture in stirred tank bioreactor provides very high volume-specific culture surface area and has been used for the production of viral vaccines (Griffiths, 1986).
  • stirred tank bioreactors have industrially been proven to be scaleable.
  • the multiplate CellcubeTM cell culture system manufactured by Corning-Costar also offers a very high volume-specific culture surface area.
  • the Cells grow on both sides of the culture plates hermetically sealed together in the shape of a compact cube.
  • the CellcubeTM culture unit is disposable. This is very desirable at the early stage production of clinical product because of the reduced capital expenditure, quality control and quality assurance costs associated with disposable systems.
  • both the stirred tank bioreactor and the CellcubeTM system were evaluated for the production of adenovirus.
  • Wave Bioreactor® is a pre-sterilized disposable bioreactor system that can be easily retrofitted with a variety of cleanroom configurations without requiring expensive CIP and SIP process utilities.
  • the Wave Bioreactor® can be a Wave Biotech® model 20/50EH.
  • the bioreactor can hold any volume of media, but in a certain embodiment the bioreactor is a 10 L (5 L working volume) bioreactor. In certain embodiments, the bioreactor can be adjusted to rock at a particular speed and angle.
  • the bioreactor may include a device for monitoring dissolved oxygen tension, such as a disposable dissolved oxygen tension (DOT) probe.
  • the bioreactor may also include a device for monitoring temperature in the media.
  • Other embodiments include a device for measuring and adjusting culture pH, such as a gas mixer which can adjust CO 2 gas percentage delivered to the media.
  • the bioreactor may or may not be a disposable bioreactor.
  • media and “medium” refers to any substance which can facilitate growth of cells.
  • the host cells are grown in media that is serum-free media.
  • the host cells are grown in media that is protein-free media.
  • a protein-free media is CD293.
  • Another example of media that can support host cell growth in a particular embodiment of the invention is DMEM+2% FBS.
  • various components and agents can be added to the media to facilitate and control cell growth.
  • the glucose concentration of the media can be maintained at a certain level. In one embodiment of the present methods for producing adenovirus, the glucose concentration is maintained between about 0.5 and about 3.0 gm glucose/liter.
  • the methods for producing an adenovirus require growing host cells in anchorage-dependent cultures, whereas other embodiments pertain to methods for producing an adenovirus in non-anchorage-dependent cultures.
  • Animal and human cells can be propagated in vitro in two modes: as non-anchorage dependent cells growing freely in suspension throughout the bulk of the culture; or as anchorage-dependent cells requiring attachment to a solid substrate for their propagation (i.e., a monolayer type of cell growth).
  • Non-anchorage dependent or suspension cultures from continuous established cell lines are the most widely used means of large scale production of cells and cell products.
  • Bioreactors are frequently employed in suspension culture systems. In the context of the present invention, however, bioreactors may also be used in anchorage-dependent cultures.
  • Large scale suspension culture can provide advantages in viral vector production. For example, the processes are relatively simple to operate and straightforward to scale up. As discussed above, homogeneous conditions can be provided in the bioreactor which allows for precise monitoring and control of culture conditions such as dissolved oxygen, and pH.
  • the bioreactors utilized in the context of selected embodiments of the present invention may be stirred tank bioreactors. Large scale suspension culture of mammalian cultures in stirred tanks have been described.
  • the instrumentation and controls for bioreactors adapted, along with the design of the fermentors, from related microbial applications. However, acknowledging the increased demand for contamination control in the slower growing mammalian cultures, improved aseptic designs were quickly implemented, improving dependability of these reactors. Instrumentation and controls are basically the same as found in other fermentors and include agitation, temperature, dissolved oxygen, and pH controls.
  • the autoanalyzer is a YSI-2700 SELECTTM analyzer.
  • stirred bioreactor Two suspension culture bioreactor designs are widely used in the industry due to their simplicity and robustness of operation—the stirred bioreactor and the airlift bioreactor.
  • the stirred bioreactor design has successfully been used on a scale of 8000 liter capacity for the production of interferon (Phillips et al., 1985; Mizrahi, 1983).
  • Cells are grown in a stainless steel tank with a height-to-diameter ratio of 1:1 to 3:1.
  • the culture is usually mixed with one or more agitators, based on bladed disks or marine propeller patterns.
  • Agitator systems offering less shear forces than blades have been described. Agitation may be driven either directly or indirectly by magnetically coupled drives. Indirect drives reduce the risk of microbial contamination through seals on stirrer shafts.
  • the bioreactor includes axial rocking of a planar platform to induce wave motions inside of the bioreactor.
  • the wave motions are induced inside of a sterilized bag made of layers of polyethylene vinyl acetate and ethyl vinyl alcohol.
  • the airlift bioreactor also initially described for microbial fermentation and later adapted for mammalian culture, relies on a gas stream to both mix and oxygenate the culture.
  • the gas stream enters a riser section of the bioreactor and drives circulation. Gas disengages at the culture surface, causing denser liquid free of gas bubbles to travel downward in the downcomer section of the bioreactor.
  • the main advantage of this design is the simplicity and lack of need for mechanical mixing. Typically, the height-to-diameter ratio is 10:1.
  • the airlift reactor scales up relatively easily, has good mass transfer of gasses and generates relatively low shear forces.
  • a batch process is a closed system in which a typical growth profile is seen. A lag phase is followed by exponential, stationary and decline phases. In such a system, the environment is continuously changing as nutrients are depleted and metabolites accumulate. This makes analysis of factors influencing cell growth and productivity, and hence optimization of the process, a complex task. Productivity of a batch process may be increased by controlled feeding of key nutrients to prolong the growth cycle. Such a fed-batch process is still a closed system because cells, products and waste products are not removed.
  • perfusion of fresh medium through the culture can be achieved by retaining the cells with a variety of devices (e.g. fine mesh spin filter, hollow fiber or flat plate membrane filters, settling tubes).
  • a true open system and the simplest perfusion process is the chemostat in which there is an inflow of medium and an outflow of cells and products.
  • Culture medium is fed to the reactor at a predetermined and constant rate which maintains the dilution rate of the culture at a value less than the maximum specific growth rate of the cells (to prevent washout of the cell mass from the reactor).
  • Culture fluid containing cells and cell products and byproducts is removed at the same rate.
  • the bioreactor system is set up to include a system to allow for media exchange.
  • filters may be incorporated into the bioreactor system to allow for separation of cells from spent media to facilitate media exchange.
  • media exchange and perfusion is conducted beginning on a certain day of cell growth. For example, media exchange and perfusion can begin on day 3 of cell growth.
  • the filter may be external to the bioreactor, or internal to the bioreactor.
  • the filter is a floating flat filter that is internal to the bioreactor.
  • the filter provides for separation between the cells and spent medium.
  • the spent culture media is withdrawn through the floating filer. Recirculation of the media may or may not be required in the various embodiments of the present invention.
  • wave action is used to minimize clogging of the filter during media perfusion.
  • the culture volume may be maintained by a load cell used to trigger fresh medium addition.
  • roller bottle Being little more than a large, differently shaped T-flask, simplicity of the system makes it very dependable and, hence, attractive. Fully automated robots are available that can handle thousands of roller bottles per day, thus eliminating the risk of contamination and inconsistency associated with the otherwise required intense human handling. With frequent media changes, roller bottle cultures can achieve cell densities of close to 0.5 ⁇ 10 6 cells/cm 2 (corresponding to approximately 10 9 cells/bottle or almost 10 7 cells/ml of culture media).
  • microcarrier cultures offer a high surface-to-volume ratio (variable by changing the carrier concentration) which leads to high cell density yields and a potential for obtaining highly concentrated cell products.
  • Cell yields are up to 1-2 ⁇ 10 7 cells/ml when cultures are propagated in a perfused reactor mode.
  • cells can be propagated in one unit process vessels instead of using many small low-productivity vessels (i.e., flasks or dishes). This results in far better nutrient utilization and a considerable saving of culture medium.
  • microcarriers settle out of suspension quickly, use of a fed-batch process or harvesting of cells can be done relatively easily.
  • microcarrier cultures are relatively easily scaled up using conventional equipment used for cultivation of microbial and animal cells in suspension.
  • One method which has shown to be particularly useful for culturing mammalian cells is microencapsulation.
  • the mammalian cells are retained inside a semipermeable hydrogel membrane.
  • a porous membrane is formed around the cells permitting the exchange of nutrients, gases, and metabolic products with the bulk medium surrounding the capsule.
  • Several methods have been developed that are gentle, rapid and non-toxic and where the resulting membrane is sufficiently porous and strong to sustain the growing cell mass throughout the term of the culture. These methods are all based on soluble alginate gelled by droplet contact with a calcium-containing solution. Lim (1982, U.S. Pat. No.
  • Microencapsulated cells are easily propagated in stirred tank reactors and, with beads sizes in the range of 150-1500 ⁇ m in diameter, are easily retained in a perfused reactor using a fine-meshed screen.
  • the ratio of capsule volume to total media volume can be maintained from as dense as 1:2 to 1:10. With intracapsular cell densities of up to 108, the effective cell density in the culture is 1-5 ⁇ 10 7 .
  • microencapsulation over other processes include the protection from the deleterious effects of shear stresses which occur from sparging and agitation, the ability to easily retain beads for the purpose of using perfused systems, scale up is relatively straightforward and the ability to use the beads for implantation.
  • Some embodiments of the current invention include cells which are anchorage-dependent in nature. 293 cells, for example, are anchorage-dependent, and when grown in suspension, the cells will attach to each other and grow in clumps, eventually suffocating cells in the inner core of each clump as they reach a size that leaves the core cells unsustainable by the culture conditions. Therefore, an efficient means of large-scale culture of anchorage-dependent cells is needed in order to effectively employ these cells to generate large quantities of adenovirus.
  • Certain embodiments of the present invention involve methods for producing an adenovirus that involve use of perfused attachment systems.
  • Perfusion refers to continuous flow at a steady rate, through or over a population of cells (of a physiological nutrient solution). It implies the retention of the cells within the culture unit as opposed to continuous-flow culture which washes the cells out with the withdrawn media (e.g., chemostat).
  • the idea of perfusion has been known since the beginning of the century, and has been applied to keep small pieces of tissue viable for extended microscopic observation. The technique was initiated to mimic the cells milieu in vivo where cells are continuously supplied with blood, lymph, or other body fluids. Without perfusion, cells in culture go through alternating phases of being fed and starved, thus limiting full expression of their growth and metabolic potential.
  • perfused culture is in response to the challenge of growing cells at high densities (i.e., 0.1-5 ⁇ 10 8 cells/ml).
  • the medium In order to increase densities beyond 2-4 ⁇ 10 6 cells/ml, the medium has to be constantly replaced with a fresh supply in order to make up for nutritional deficiencies and to remove toxic products.
  • Perfusion allows for a far better control of the culture environment (pH, pO 2 , nutrient levels, etc.) and is a means of significantly increasing the utilization of the surface area within a culture for cell attachment.
  • this reactor comprises an improved reactor for culturing of both anchorage- and non-anchorage-dependent cells.
  • the reactor is designed as a packed bed with a means to provide internal recirculation.
  • a fiber matrix carrier is placed in a basket within the reactor vessel. A top and bottom portion of the basket has holes, allowing the medium to flow through the basket.
  • a specially designed impeller provides recirculation of the medium through the space occupied by the fiber matrix for assuring a uniform supply of nutrient and the removal of wastes. This simultaneously assures that a negligible amount of the total cell mass is suspended in the medium.
  • the combination of the basket and the recirculation also provides a bubble-free flow of oxygenated medium through the fiber matrix.
  • the fiber matrix is a non-woven fabric having a “pore” diameter of from 10 ⁇ m to 100 ⁇ m, providing for a high internal volume with pore volumes corresponding to 1 to 20 times the volumes of individual cells.
  • the CellcubeTM (Corning-Costar) module provides a large styrenic surface area for the immobilization and growth of substrate attached cells. It is an integrally encapsulated sterile single-use device that has a series of parallel culture plate joined to create thin sealed laminar flow spaces between adjacent plates.
  • the CellcubeTM module has inlet and outlet ports that are diagonally opposite each other and help regulate the flow of media.
  • the amount of time between the initial seeding and the start of the media perfusion is dependent on the density of cells in the seeding inoculum and the cell growth rate.
  • the measurement of nutrient concentration in the circulating media is a good indicator of the status of the culture.
  • Cells within the system reach a higher density of solution (cells/ml) than in traditional culture systems.
  • Many typically used basal media are designed to support 1-2 ⁇ 10 6 cells/ml/day.
  • a typical CellcubeTM run with an 85,000 cm 2 surface, contains approximately 6 L media within the module. The cell density often exceeds 10 7 cells/mL in the culture vessel. At confluence, 2-4 reactor volumes of media are required per day.
  • the timing and parameters of the production phase of cultures depends on the type and use of a particular cell line. Many cultures require a different media for production than is required for the growth phase of the culture. The transition from one phase to the other will likely require multiple washing steps in traditional cultures.
  • the CellcubeTM system employs a perfusion system. On of the benefits of such a system is the ability to provide a gentle transition between various operating phases. The perfusion system negates the need for traditional wash steps that seek to remove serum components in a growth medium.
  • adenovirus is produced from anchorage-dependent culture of host cells (e.g., HEK293 cells). Scale-up of adenoviral vector production is constrained by the anchorage-dependency of 293A cells. To facilitate scale-up and meet future demand for adenoviral vectors, significant efforts have been devoted to the development of alternative production processes that are amenable to scale-up. Methods include growing HEK293 cells in bioreactors and adaptation of host cells into suspension cultures.
  • the media used in the methods for producing an adenovirus is a serum-free media.
  • the media is a protein-free media.
  • bioreactors may be adapted for serum-free suspension culture of cells. Filtration of media with media exchange may or may not be included in the system.
  • the present invention pertains to methods of producing an adenovirus that include infecting the host cells with an adenovirus. Typically, the virus will simply be exposed to the appropriate host cell under physiologic conditions, permitting uptake of the virus. One of skill in the art would be familiar with the wide range of techniques available for initiating virus infection.
  • the temperature at which infection of the host cells is performed is 37° C.
  • the infection temperature is at temperature that is less than 37° C. This is based on the inventors' discovery that infection temperatures less than 37° C. provide for optimal production of adenovirus.
  • the temperature may be 32.1° C., 32.2° C., 32.3° C., 32.4° C., 32.5° C., 32.6° C., 32.7° C., 32.8° C., 32.9° C., 33.0° C., 33.1° C., 33.2° C., 33.3° C., 33.4° C., 33.5° C., 33.6° C., 33.7° C., 33.8° C., 33.9° C., 34.0° C., 34.1° C., 34.2° C., 34.3° C., 34.4° C., 34.5° C., 34.6° C., 34.7° C., 34.8° C., 34.9° C., 35.0° C., 35.1° C., 35.2° C., 35.3° C., 35.4° C., 35.5° C., 35.6° C., 35.7° C., 35.8° C., 35.9° C., 36.0° C., 36.1°
  • one convenient way to measure temperature would be to use a real time digital device to measure the temperature inside an incubator. Prior to the procedure, the digital device can be calibrated using traceable temperature calibration equipment to verify accuracy of the digital device.
  • the methods for producing an adenovirus may involve initiating virus infection by diluting the host cells with fresh media and adenovirus. This avoids the need for a separate medium exchange step prior to infection.
  • the invention contemplates that any amount of dilution of the host cells is contemplated by the present invention. In a certain embodiment, the host cells are diluted 10-fold with fresh media.
  • the invention also contemplates any amount of virus added to initiate infection. However, in a certain embodiment of the present invention, virus infection will be initiated by adding 50 vp/host cell.
  • virus infection can be allowed to proceed for any length of time. However, in a certain embodiment, virus infection is allowed to proceed for 4 days. In another embodiment of the present invention, host cell growth is allowed to occur in one bioreactor, and infection of host cells is conducted in a second bioreactor.
  • the term “adenovirus preparation” will be used herein to describe the reaction mixture following initiation of infection with adenovirus.
  • the adenovirus preparation may include host cells that have undergone lysis, cell fragments, adenovirus, media, and any other components present in the reaction mixture during infection.
  • the adenovirus preparation may include intact host cells, depending on how long infection was allowed to proceed. Some or all of the host cells may have undergone cell lysis, with release of viral particles into the surrounding media.
  • adenovirus isolation will occur at any time and by any means known to those of skill in the art following infection. For example, in one embodiment of the present invention, isolating the adenovirus from the adenovirus preparation occurs 4 days after viral infection is completed.
  • a recombinant adenovirus is contemplated for the delivery of expression constructs.
  • “Recombinant adenovirus,” “adenovirus vector” or “adenoviral expression vector” is meant to include those constructs containing adenovirus sequences sufficient to (a) support packaging of the construct and (b) to ultimately express a tissue or cell-specific construct that has been cloned therein.
  • the recombinant adenovirus may encode a recombinant gene.
  • a recombinant adenovirus may include any of the engineered vectors that comprise adenoviral sequences.
  • An adenovirus expression vector according to the present invention comprises a genetically engineered form of the adenovirus.
  • the nature of the adenovirus vector is not believed to be crucial to the successful practice of the invention.
  • the adenovirus may be of any of the known serotypes and/or subgroups A-F.
  • Adenovirus type 5 of subgroup C is the preferred starting material in order to obtain one adenovirus vector for use in the present invention. This is because adenovirus type 5 is a human adenovirus about which a great deal of biochemical and genetic information is known, and it has historically been used for most constructions employing adenovirus as a vector.
  • adenoviral gene transfer includes the ability to infect a wide variety of cell types, including non-dividing cells, a mid-sized genome, ease of manipulation, high infectivity and they can be grown to high titers (Wilson, 1996). Further, adenoviral infection of host cells does not result in chromosomal integration because adenoviral DNA can replicate in an episomal manner, without potential genotoxicity associated with other viral vectors. Adenoviruses also are structurally stable (Marienfeld et al., 1999) and no genome rearrangement has been detected after extensive amplification (Parks et al., 1997; Bett et al., 1993).
  • Adenovirus growth and manipulation is known to those of skill in the art, and exhibits broad host range in vitro and in vivo (U.S. Pat. No. 5,670,488; U.S. Pat. No. 5,932,210; U.S. Pat. No. 5,824,544).
  • This group of viruses can be obtained in high titers, e.g., 10 9 to 10 11 plaque-forming units per ml, and they are highly infective.
  • the life cycle of adenovirus does not require integration into the host cell genome.
  • the foreign genes delivered by adenovirus vectors are episomal and, therefore, have low genotoxicity to host cells.
  • adenovirus based vectors offer several unique advantages over other vector systems, they often are limited by vector immunogenicity, size constraints for insertion of recombinant genes, low levels of replication, and low levels of transgene expression.
  • a major concern in using adenoviral vectors is the generation of a replication-competent virus during vector production in a packaging cell line or during gene therapy treatment of an individual. The generation of a replication-competent virus could pose serious threat of an unintended viral infection and pathological consequences for the patient.
  • Certain embodiments of the present invention pertain to methods of producing an adenovirus that involve replication-deficient adenovirus.
  • Armentano et al. describe the preparation of a replication-deficient adenovirus vector, claimed to eliminate the potential for the inadvertent generation of a replication-competent adenovirus (U.S. Pat. No. 5,824,544).
  • the replication-deficient adenovirus method comprises a deleted E1 region and a relocated protein IX gene, wherein the vector expresses a heterologous, mammalian gene.
  • a common approach for generating adenoviruses for use as a gene transfer vector is the deletion of the E1 gene (E1 ⁇ ), which is involved in the induction of the E2, E3 and E4 promoters (Graham and Prevec, 1995). Subsequently, a therapeutic gene or genes can be inserted recombinantly in place of the E1 gene, wherein expression of the therapeutic gene(s) is driven by the E1 promoter or a heterologous promoter. The E1-, replication-deficient virus is then proliferated in a “helper” cell line that provides the E1 polypeptides in trans (e.g., the human embryonic kidney cell line 293).
  • a “helper” cell line that provides the E1 polypeptides in trans
  • the E3 region, portions of the E4 region or both may be deleted, wherein a heterologous nucleic acid sequence under the control of a promoter operable in eukaryotic cells is inserted into the adenovirus genome for use in gene transfer (U.S. Pat. No. 5,670,488; U.S. Pat. No. 5,932,210).
  • the invention may include methods of producing an adenovirus where the adenovirus is a recombinant adenovirus encoding a recombinant gene.
  • the recombinant gene may be operatively linked to a promoter.
  • the recombinant gene is a therapeutic gene.
  • the invention contemplates use of any gene that has therapeutic or potential therapeutic value in the treatment of a disease or genetic disorder.
  • One of skill in the art would be familiar with the wide range of such genes that have been identified.
  • Gene therapy generally involves the introduction into cells of therapeutic genes, also known as transgenes, whose expression results in amelioration or treatment of disease or genetic disorders.
  • the therapeutic genes involved may be those that encode proteins, structural or enzymatic RNAs, inhibitory products such as antisense RNA or DNA, or any other gene product. Expression is the generation of such a gene product or the resultant effects of the generation of such a gene product. Thus, enhanced expression includes the greater production of any therapeutic gene or the augmentation of that product's role in determining the condition of the cell, tissue, organ or organism.
  • the delivery of therapeutic genes by adenoviral vectors involves what may be termed transduction of cells. As used here, transduction is defined as the introduction into a cell a therapeutic gene, transgene, or transgene construct by an adenoviral or related vector.
  • adenoviral gene delivery-based gene therapies are being developed for liver diseases (Han et al., 1999), psychiatric diseases (Lesch, 1999), neurological diseases (Hermens and Verhaagen, 1998), coronary diseases (Feldman et al., 1996), muscular diseases (Petrof, 1998), and various cancers such as colorectal (Dorai et al, 1999), bladder (Irie et al., 1999), prostate (Mincheff et al., 2000), head and neck (Blackwell et al., 1999), breast (Stewart et al., 1999), lung (Batra et al., 1999) and ovarian (Vanderkwaak et al., 1999).
  • the particular therapeutic gene encoded by the adenoviral vector is not limiting and includes those useful for various therapeutic and research purposes, as well as reporter genes and reporter gene systems and contructs useful in tracking the expression of transgenes and the effectiveness of adenoviral and adenoviral vector transduction.
  • reporter genes and reporter gene systems and contructs useful in tracking the expression of transgenes and the effectiveness of adenoviral and adenoviral vector transduction.
  • developmental genes e.g.
  • adhesion molecules cyclin kinase inhibitors, Wnt family members, Pax family members, Winged helix family members, Hox family members, cytokines/lymphokines and their receptors, growth or differentiation factors and their receptors, neurotransmitters and their receptors), oncogenes (e.g.
  • ABLI BLC1, BCL6, CBFA1, CBL, CSFIR, ERBA, ERBB, EBRB2, ETS1, ETS1, ETV6, FGR, FOX, FYN, HCR, HRAS, JUN, KRAS, LCK, LYN, MDM2, MLL, MYB, MYC, MYCLI, MYCN, NRAS, PIM1, PML, RET, SRC, TAL1, TCL3 and YES), tumor suppresser genes (e.g. APC, BRCA1, BRCA2, MADH4, MCC, NF1, NF2, RB1, TP53 and WT1), enzymes (e.g.
  • ACP desaturases and hycroxylases, ADP-glucose pyrophorylases, ATPases, alcohol dehycrogenases, amylases, amyloglucosidases, catalases, cellulases, cyclooxygenases, decarboxylases, dextrinases, esterases, DNA and RNA polymerases, hyaluron synthases, galactosidases, glucanases, glucose oxidases, GTPases, helicases, hemicellulases, hyaluronidases, integrases, invertases, isomersases, kinases, lactases, lipases, lipoxygenases, lyases, lysozymes, pectinesterases, peroxidases, phosphatases, phospholipases, phophorylases, polygalacturonases, proteinases and
  • the therapeutic gene can also be an antisense gene or sequence whose expression in the target cell enables the expression of cellular genes or the transcription of cellular mRNA to be controlled, or instance ribozymes. Such sequence can, for example, be transcribed in the target cell into RNAs complementary to cellular mRNAs.
  • the therapeutic gene can also be a gene coding for an antigenic peptide capable of generating an immune response in man. In this particular embodiment, the invention hence makes it possible to produce vaccines enabling humans to be immunized, in particular against microorganisms and viruses.
  • the tumor suppressor oncogenes function to inhibit excessive cellular proliferation.
  • the inactivation of these genes destroys their inhibitory activity, resulting in unregulated proliferation.
  • the tumor suppressors p53, p16 and C-CAM are described below.
  • p53 currently is recognized as a tumor suppressor gene. High levels of mutant p53 have been found in many cells transformed by chemical carcinogenesis, ultraviolet radiation, and several viruses, including SV40.
  • the p53 gene is a frequent target of mutational inactivation in a wide variety of human tumors and is already documented to be the most frequently-mutated gene in common human cancers. It is mutated in over 50% of human NSCLC (Holistein et al., 1991) and in a wide spectrum of other tumors.
  • the p53 gene encodes a 393-amino-acid phophoprotein that can form complexes with host proteins such as large-T antigen and E1B.
  • the protein is found in normal tissues and cells, but at concentrations which are minute by comparison with transformed cells or tumor tissue.
  • wild-type p53 appears to be important in regulating cell growth and division. Overexpression of wild-type p53 has been shown in some cases to be anti-proliferative in human tumor cell lines. Thus, p53 can act as a negative regulator of cell growth (Weinberg, 1991) and may directly suppress uncontrolled cell growth or indirectly activate genes that suppress this growth. Thus, absence or inactivation of wild-type p53 may contribute to transformation. However, some studies indicate that the presence of mutant p53 may be necessary for full expression of the transforming potential of the gene.
  • Wild-type p53 is recognized as an important growth regulator in many cell types. Missense mutations are common for the p53 gene and are essential for the transforming ability of the oncogene. A single genetic change prompted by point mutations can create carcinogenic p53. Unlike other oncogenes, however, p53 point mutations are know to occur in at least 30 distinct codons, often creating dominant alleles that produce shifts in cell phenotype without a reduction to homozygosity. Additionally, many of these dominant negative alleles appear to be tolerated in the organism and passed on in the germ line. Various mutant alleles appear to range from minimally dysfunctional to strongly penetrant, dominant negative alleles (Weinberg, 1991).
  • tumor suppressors that may be employed according to the present invention include BRCATM, BRCA2, zac1, p73, MMAC-1, ATM, HIC-1, DPC-4, FHIT, NF2, APC, DCC, PTEN, ING1, NOEY1, NOEY2, PML, OVCA1, MADR2, WT1, 53BP2, and IRF-1.
  • genes that may be employed according to the present invention include Rb, APC, DCC, NF-1, NF-2, WT-1, MEN-I, MEN-II, zac1, p73, VHL, MMAC1/PTEN, DBCCR-1, FCC, rsk-3, p27, p27/p16 fusions, p21/p27 fusions, anti-thrombotic genes (e.g., COX-1, TFPI), PGS, Dp, E2F, ras, myc, neu, raf, erb, fms, trk, ret, gsp, hst, abl, E1A, p300, genes involved in angiogenesis (e.g., VEGF, FGF, thrombospondin, BAI-1, GDAIF, or their receptors) and MCC.
  • angiogenesis e.g., VEGF, FGF, thrombospondin, BAI-1,
  • the adenovirus comprises an exogenous gene construct that is an mda-7 gene.
  • MDA-7 is another putative tumor suppressor that has been shown to suppress the growth of cancer cells that are p53-wild-type, p53-null and p53-mutant.
  • the observed upregulation of the apoptosis-related Bax gene in p53 null cells indicates that MDA-7 is capable of using p53-independent mechanisms to induce the destruction of cancer cells.
  • a colony inhibition assay was used to demonstrate that elevated expression of MDA-7 enhanced growth inhibition in human cervical carcinoma (HeLa), human breast carcinoma (MCF-7 and T47D), colon carcinoma (LS174T and SW480), nasopharyngeal carcinoma (HONE-1), prostate carcinoma (DU-145), melanoma (HO-1 and C8161), glioblastome multiforme (GBM-18 and T98G), and osteosarcoma (Saos-2).
  • MDA-7 overexpression in normal cells HMECs, HBL-100, and CREF-Trans6
  • the data indicates that growth inhibition by elevated expression of MDA-7 is more effective in vitro in cancer cells than in normal cells.
  • MDA-7 has broad therapeutic, prognostic and diagnostic potential as an inducer of PKR and, consequently, an enhancer of an induced immune response.
  • genes are also considered therapeutic genes.
  • Particularly appropriate genes for expression include those genes that are thought to be expressed at less than normal level in the target cells of the subject mammal. Examples of particularly useful gene products include carbamoyl synthetase I, omithine transcarbamylase, arginosuccinate synthetase, arginosuccinate lyase, and arginase.
  • Other desirable gene products include fumarylacetoacetate hydrolase, phenylalanine hydroxylase, alpha-1 antitrypsin, glucose-6-phosphatase, low-density-lipoprotein receptor, porphobilinogen deaminase, factor VIII, factor IX, cystathione .beta.-synthase, branched chain ketoacid decarboxylase, albumin, isovaleryl-CoA dehydrogenase, propionyl CoA carboxylase, methyl malonyl CoA mutase, glutaryl CoA dehydrogenase, insulin, .beta.-glucosidase, pyruvate carboxylase, hepatic phosphorylase, phosphorylase kinase, glycine decarboxylase (also referred to as P-protein), H-protein, T-protein, Menkes disease copper-transporting ATPase, and Wilson's disease copper-transporting ATPase
  • gene products include cytosine deaminase, hypoxanthine-guanine phosphoribosyltransferase, galactose-1-phosphate uridyltransferase, phenylalanine hydroxylase, glucocerbrosidase, sphingomyelinase, ⁇ -L-iduronidase, glucose-6-phosphate dehydrogenase, HSV thymidine kinase and human thymidine kinase.
  • Hormones are another group of genes that may be used in the vectors described herein.
  • growth hormone prolactin, placental lactogen, luteinizing hormone, follicle-stimulating hormone, chorionic gonadotropin, thyroid-stimulating hormone, leptin, adrenocorticotropin (ACTH), angiotensin I and II, ⁇ -endorphin, ⁇ -melanocyte stimulating hormone ( ⁇ -MSH), cholecystokinin, endothelin I, galanin, gastric inhibitory peptide (GIP), glucagon, insulin, lipotropins, neurophysins, somatostatin, calcitonin, calcitonin gene related peptide (CGRP), ⁇ -calcitonin gene related peptide, hypercalcemia of malignancy factor (1-40), parathyroid hormone-related protein (107-139) (PTH-rP), parathyroid hormone-related protein (107-111) (PTH-rP), glucagon-like peptide (GLP-1), pancreastatin, pan
  • Examples of diseases for which the present viral vector would be useful include, but are not limited to, adenosine deaminase deficiency, human blood clotting factor IX deficiency in hemophilia B, and cystic fibrosis, which would involve the replacement of the cystic fibrosis conductance regulator gene.
  • the vectors embodied in the present invention could also be used for treatment of hyperproliferative disorders such as rheumatoid arthritis or restenosis by transfer of genes encoding angiogenesis inhibitors or cell cycle inhibitors. Transfer of prodrug activators such as the HSV-TK gene can be also be used in the treatment of hyperploiferative disorders, including cancer.
  • Oncogenes such as ras, myc, neu, raf erb, src, fms, jun, trk, ret, gsp, hst, bcl and abl also are suitable targets. However, for therapeutic benefit, these oncogenes would be expressed as an antisense nucleic acid, so as to inhibit the expression of the oncogene.
  • antisense nucleic acid is intended to refer to the oligonucleotides complementary to the base sequences of oncogene-encoding DNA and RNA.
  • Antisense oligonucleotides when introduced into a target cell, specifically bind to their target nucleic acid and interfere with transcription, RNA processing, transport and/or translation. Targeting double-stranded (ds) DNA with oligonucleotide leads to triple-helix formation; targeting RNA will lead to double-helix formation.
  • ds double-stranded
  • Antisense constructs may be designed to bind to the promoter and other control regions, exons, introns or even exon-intron boundaries of a gene.
  • Antisense RNA constructs, or DNA encoding such antisense RNAs may be employed to inhibit gene transcription or translation or both within a host cell, either in vitro or in vivo, such as within a host animal, including a human subject.
  • Nucleic acid sequences comprising “complementary nucleotides” are those which are capable of base-pairing according to the standard Watson-Crick complementarity rules.
  • the larger purines will base pair with the smaller pyrimidines to form only combinations of guanine paired with cytosine (G:C) and adenine paired with either thymine (A:T), in the case of DNA, or adenine paired with uracil (A:U) in the case of RNA.
  • complementary or antisense sequences mean nucleic acid sequences that are substantially complementary over their entire length and have very few base mismatches. For example, nucleic acid sequences of fifteen bases in length may be termed complementary when they have a complementary nucleotide at thirteen or fourteen positions with only single or double mismatches. Naturally, nucleic acid sequences which are “completely complementary” will be nucleic acid sequences which are entirely complementary throughout their entire length and have no base mismatches.
  • any sequence 17 bases long should occur only once in the human genome and, therefore, suffice to specify a unique target sequence.
  • shorter oligomers are easier to make and increase in vivo accessibility, numerous other factors are involved in determining the specificity of hybridization. Both binding affinity and sequence specificity of an oligonucleotide to its complementary target increases with increasing length. It is contemplated that oligonucleotides of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more base pairs will be used.
  • One can readily determine whether a given antisense nucleic acid is effective at targeting of the corresponding host cell gene simply by testing the constructs in vitro to determine whether the endogenous gene's function is affected or whether the expression of related genes having complementary sequences is affected.
  • antisense constructs which include other elements, for example, those which include C-5 propyne pyrimidines.
  • Oligonucleotides which contain C-5 propyne analogues of uridine and cytidine have been shown to bind RNA with high affinity and to be potent antisense inhibitors of gene expression (Wagner et al., 1993).
  • ribozyme refers to an RNA-based enzyme capable of targeting and cleaving particular base sequences in oncogene DNA and RNA. Ribozymes can either be targeted directly to cells, in the form of RNA oligo-nucleotides incorporating ribozyme sequences, or introduced into the cell as an expression construct encoding the desired ribozymal RNA. Ribozymes may be used and applied in much the same way as described for antisense nucleic acids.
  • Other therapeutics genes might include genes encoding antigens such as viral antigens, bacterial antigens, fungal antigens or parasitic antigens.
  • Viruses include picornavirus, coronavirus, togavirus, flavirvirus, rhabdovirus, paramyxovirus, orthomyxovirus, bunyavirus, arenvirus, reovirus, retrovirus, papovavirus, parvovirus, herpesvirus, poxvirus, hepadnavirus, and spongiform virus.
  • Preferred viral targets include influenza, herpes simplex virus 1 and 2, measles, small pox, polio or HIV.
  • Pathogens include trypanosomes, tapeworms, roundworms, helminths.
  • tumor markers such as fetal antigen or prostate specific antigen
  • Preferred examples include HIV env proteins and hepatitis B surface antigen.
  • Administration of a vector according to the present invention for vaccination purposes would require that the vector-associated antigens be sufficiently non-immunogenic to enable long term expression of the transgene, for which a strong immune response would be desired.
  • vaccination of an individual would only be required infrequently, such as yearly or biennially, and provide long term immunologic protection against the infectious agent.
  • the polynucleotide encoding the therapeutic gene will be under the transcriptional control of a promoter and a polyadenylation signal. Therefore, certain embodiments of the present invention involve methods for producing an adenovirus wherein the adenovirus comprises an adenoviral vector encoding an exogenous gene construct that is operatively linked to a promoter.
  • a “promoter” refers to a DNA sequence recognized by the synthetic machinery of the host cell, or introduced synthetic machinery, that is required to initiate the specific transcription of a gene.
  • a polyadenylation signal refers to a DNA sequence recognized by the synthetic machinery of the host cell, or introduced synthetic machinery, that is required to direct the addition of a series of nucleotides on the end of the mRNA transcript for proper processing and trafficking of the transcript out of the nucleus into the cytoplasm for translation.
  • the phrases “operatively linked,” “under control,” and “under transcriptional control” mean that the promoter is in the correct location in relation to the polynucleotide to control RNA polymerase initiation and expression of the polynucleotide.
  • promoter will be used here to refer to a group of transcriptional control modules that are clustered around the initiation site for RNA polymerase II. Much of the thinking about how promoters are organized derives from analyses of several viral promoters, including those for the HSV thymidine kinase (tk) and SV40 early transcription units. These studies, augmented by more recent work, have shown that promoters are composed of discrete functional modules, each consisting of approximately 7-20 bp of DNA, and containing one or more recognition sites for transcriptional activator or repressor proteins.
  • At least one module in each promoter functions to position the start site for RNA synthesis.
  • the best known example of this is the TATA box, but in some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation.
  • Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well.
  • the spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the tk promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription.
  • the particular promoter that is employed to control the expression of a therapeutic gene is not believed to be critical, so long as it is capable of expressing the polynucleotide in the targeted cell.
  • the promoter may be a tissue-specific promoter or an inducible promoter. Examples of promoters that may be employed include SV40 E1, RSV LTR, ⁇ -actin, CMV-IE, adenovirus major late, polyoma F9-1, ⁇ -fetal protein promoter, egr-1, or tyrosinase promoter.
  • SV40 E1 SV40 E1
  • RSV LTR ⁇ -actin
  • CMV-IE adenovirus major late
  • polyoma F9-1 polyoma F9-1
  • ⁇ -fetal protein promoter egr-1
  • tyrosinase promoter tyrosinase promoter.
  • a human cell it is preferable to position the polynucleotide coding region adjacent to and under the control of a promoter that is capable of being expressed in a human cell.
  • a promoter might include either a human or viral promoter.
  • a list of promoters is provided in the Table 1.
  • the promoter may be a constitutive promoter, an inducible promoter, or a tissue-specific promoter.
  • An inducible promoter is a promoter which is inactive or exhibits low activity except in the presence of an inducer substance.
  • Some examples of promoters that may be included as a part of the present invention include, but are not limited to, MT II, MMTV, Collagenase, Stromelysin, SV40, Murine MX gene, ⁇ -2-Macroglobulin, MHC class I gene h-2 kb, HSP70, Proliferin, Tumor Necrosis Factor, or Thyroid Stimulating Hormone ⁇ gene.
  • the associated inducers are shown in Table 2.
  • a promoter that is “endogenous” or “constitutive” is a promoter that is one naturally associated with a gene or sequence, as may be obtained by isolating the 5′ non-coding sequences located upstream of the coding segment and/or exon.
  • TPA Phorbol Ester
  • TAA Heavy metals MMTV (mouse mammary tumor Glucocorticoids virus) ⁇ -Interferon poly(rI)X poly(rc) Adenovirus 5 E2 Ela c-jun Phorbol Ester (TPA), H 2 O 2 Collagenase Phorbol Ester (TPA) Stromelysin Phorbol Ester (TPA), IL-1 SV40 Phorbol Ester (TPA) Murine MX Gene Interferon, Newcastle Disease Virus GRP78 Gene A23187 ⁇ -2-Macroglobulin IL-6 Vimentin Serum MHC Class I Gene H-2 kB Interferon HSP70 Ela, SV40 Large T Antigen Proliferin Phorbol Ester-TPA Tumor Necrosis Factor FMA Thyroid Stimulating Hormone ⁇ Thyroid Hormone Gene
  • the human cytomegalovirus (CMV) immediate early gene promoter, the SV40 early promoter and the Rous sarcoma virus long terminal repeat can be used to obtain high-level expression of the polynucleotide of interest.
  • CMV cytomegalovirus
  • the use of other viral or mammalian cellular or bacterial phage promoters which are well-known in the art to achieve expression of polynucleotides is contemplated as well, provided that the levels of expression are sufficient to produce a growth inhibitory effect.
  • a promoter with well-known properties, the level and pattern of expression of a polynucleotide following transfection can be optimized. For example, selection of a promoter which is active in specific cells, such as tyrosinase (melanoma), alpha-fetoprotein and albumin (liver tumors), CC10 (lung tumor) and prostate-specific antigen (prostate tumor) will permit tissue-specific expression of the therapeutic gene.
  • a promoter which is active in specific cells such as tyrosinase (melanoma), alpha-fetoprotein and albumin (liver tumors), CC10 (lung tumor) and prostate-specific antigen (prostate tumor) will permit tissue-specific expression of the therapeutic gene.
  • Enhancers were originally detected as genetic elements that increased transcription from a promoter located at a distant position on the same molecule of DNA. This ability to act over a large distance had little precedent in classic studies of prokaryotic transcriptional regulation. Subsequent work showed that regions of DNA with enhancer activity are organized much like promoters. That is, they are composed of many individual elements, each of which binds to one or more transcriptional proteins.
  • enhancers The basic distinction between enhancers and promoters is operational. An enhancer region as a whole must be able to stimulate transcription at a distance; this need not be true of a promoter region or its component elements. On the other hand, a promoter must have one or more elements that direct initiation of RNA synthesis at a particular site and in a particular orientation, whereas enhancers lack these specificities. Promoters and enhancers are often overlapping and contiguous, often seeming to have a very similar modular organization.
  • Eukaryotic Promoter Data Base Eukaryotic Promoter Data Base
  • Eukaryotic cells can support cytoplasmic transcription from certain bacteriophage promoters if the appropriate bacteriophage polymerase is provided, either as part of the delivery complex or as an additional genetic expression vector.
  • polyadenylation signal to effect proper polyadenylation of the gene transcript.
  • the nature of the polyadenylation signal is not believed to be crucial to the successful practice of the invention, and any such sequence may be employed.
  • polyadenylation signals as that from SV40, bovine growth hormone, and the herpes simplex virus thymidine kinase gene have been found to function well in a number of target cells.
  • Adenoviral infection results in the lysis of the cells being infected.
  • the lytic characteristics of adenovirus infection permit two different modes of virus isolation and production. One is harvesting infected cells prior to cell lysis. The other mode is harvesting virus supernatant after complete cell lysis by the produced virus. For the latter mode, longer incubation times are required in order to achieve complete cell lysis. This prolonged incubation time after virus infection creates a serious concern about increased possibility of generation of replication competent adenovirus (RCA), particularly for the current first generation adenoviral vectors (E1-deleted vector). Therefore, in certain embodiments of the present invention, the methods for producing an adenovirus involve harvesting the host cells and then lysing the host cells.
  • RCA replication competent adenovirus
  • Table 3 lists the most common methods that have been used for lysing cells after cell harvest. TABLE 3 Methods used for cell lysis Methods Procedures Comments Freeze-thaw Cycling between dry ice Easy to carry out at lab and 37° C. water bath scale. High cell lysis efficiency Not scaleable Not recommended for large scale manufacturing Solid Shear French Press Capital equipment Hughes Press investment Virus containment concerns Lack of experience Detergent lysis Non-ionic detergent Easy to carry out at both lab solutions such as Tween, and manufacturing Triton, NP-40, etc.
  • the methods for producing an adenovirus involve isolating the adenovirus by lysing the host cells with a detergent.
  • Cells are bounded by membranes. In order to release components of the cell, it is necessary to break open the cells.
  • the most advantageous way in which this can be accomplished, according to the present invention, is to solubilize the membranes with the use of detergents.
  • Detergents are amphipathic molecules with an apolar end of aliphatic or aromatic nature and a polar end which may be charged or uncharged. Detergents are more hydrophilic than lipids and thus have greater water solubility than lipids. They allow for the dispersion of water insoluble compounds into aqueous media and are used to isolate and purify proteins in a native form.
  • any detergent capable of lysing the host cells is contemplated by the claimed invention.
  • One of skill in the art would be familiar with the wide range of detergents available for lysing cells.
  • Detergents can be denaturing or non-denaturing.
  • the former can be anionic such as sodium dodecyl sulfate or cationic such as ethyl trimethyl ammonium bromide. These detergents totally disrupt membranes and denature the protein by breaking protein-protein interactions.
  • Non denaturing detergents can be divided into non-anionic detergents such as Triton®X-100, bile salts such as cholates and zwitterionic detergents such as CHAPS. Zwitterionics contain both cationic and anion groups in the same molecule, the positive electric charge is neutralized by the negative charge on the same or adjacent molecule.
  • Denaturing agents such as SDS bind to proteins as monomers and the reaction is equilibrium driven until saturated. Thus, the free concentration of monomers determines the necessary detergent concentration.
  • SDS binding is cooperative i.e. the binding of one molecule of SDS increase the probability of another molecule binding to that protein, and alters proteins into rods whose length is proportional to their molecular weight.
  • Non-denaturing agents such as Triton®X-100 do not bind to native conformations nor do they have a cooperative binding mechanism. These detergents have rigid and bulky apolar moieties that do not penetrate into water soluble proteins. They bind to the hydrophobic parts of proteins. Triton®X100 and other polyoxyethylene nonanionic detergents are inefficient in breaking protein-protein interaction and can cause artifactual aggregations of protein. These detergents will, however, disrupt protein-lipid interactions but are much gentler and capable of maintaining the native form and functional capabilities of the proteins.
  • Dialysis works well with detergents that exist as monomers. Dialysis is somewhat ineffective with detergents that readily aggregate to form micelles because the micelles are too large to pass through dialysis. Ion exchange chromatography can be utilized to circumvent this problem. The disrupted protein solution is applied to an ion exchange chromatography column and the column is then washed with buffer minus detergent. The detergent will be removed as a result of the equilibration of the buffer with the detergent solution. Alternatively the protein solution may be passed through a density gradient. As the protein sediments through the gradients the detergent will come off due to the chemical potential.
  • a single detergent is not versatile enough for the solubilization and analysis of the milieu of proteins found in a cell.
  • the proteins can be solubilized in one detergent and then placed in another suitable detergent for protein analysis.
  • the protein detergent micelles formed in the first step should separate from pure detergent micelles. When these are added to an excess of the detergent for analysis, the protein is found in micelles with both detergents. Separation of the detergent-protein micelles can be accomplished with ion exchange or gel filtration chromatography, dialysis or buoyant density type separations.
  • Triton®X-100, X114 and NP-40 have the same basic characteristics but are different in their specific hydrophobic-hydrophilic nature. All of these heterogeneous detergents have a branched 8-carbon chain attached to an aromatic ring. This portion of the molecule contributes most of the hydrophobic nature of the detergent. Triton®X detergents are used to solublize membrane proteins under non-denaturing conditions. The choice of detergent to solubilize proteins will depend on the hydrophobic nature of the protein to be solubilized. Hydrophobic proteins require hydrophobic detergents to effectively solubilize them.
  • Triton®X-100 and NP-40 are very similar in structure and hydrophobicity and are interchangeable in most applications including cell lysis, delipidation protein dissociation and membrane protein and lipid solubilization. Generally 2 mg detergent is used to solubilize 1 mg membrane protein or 10 mg detergent/1 mg of lipid membrane. Triton®X-114 is useful for separating hydrophobic from hydrophilic proteins.
  • Triton®X detergents are similar in structure to Triton®X detergents in that they have varying lengths of polyoxyethylene chains attached to a hydrophobic chain. However, unlike Triton®X detergents, the Brij detergents do not have an aromatic ring and the length of the carbon chains can vary. The Brij® detergents are difficult to remove from solution using dialysis but may be removed by detergent removing gels. Brij®58 is most similar to Triton®X100 in its hydrophobic/hydrophilic characteristics. Brij®-35 is a commonly used detergent in HPLC applications.
  • ⁇ -Octyl- ⁇ -D-glucoside octylglucopyranoside
  • ⁇ -Octyl- ⁇ -D-thioglucoside octylthioglucopyranoside, OTG
  • Octylthioglucoside was first synthesized to offer an alternative to octylglucoside. Octylglucoside is expensive to manufacture and there are some inherent problems in biological systems because it can be hydrolyzed by ⁇ -glucosidase.
  • the Tween detergents are nondenaturing, nonionic detergents. They are polyoxyethylene sorbitan esters of fatty acids. Tween® 20 and Tween 80 detergents are used as blocking agents in biochemical applications and are usually added to protein solutions to prevent nonspecific binding to hydrophobic materials such as plastics or nitrocellulose. They have been used as blocking agents in ELISA and blotting applications. Generally, these detergents are used at concentrations of 0.01-1.0% to prevent nonspecific binding to hydrophobic materials.
  • Tween® 20 and other nonionic detergents have been shown to remove some proteins from the surface of nitrocellulose.
  • Tween® 80 has been used to solubilize membrane proteins, present nonspecific binding of protein to multiwell plastic tissue culture plates and to reduce nonspecific binding by serum proteins and biotinylated protein A to polystyrene plates in ELISA.
  • Tween® 80 is derived from oleic acid with a C 18 chain while Tween® 20 is derived from lauric acid with a C 12 chain.
  • the longer fatty acid chain makes the Tween® 80 detergent less hydrophilic than Tween® 20 detergent. Both detergents are very soluble in water.
  • Tween® detergents are difficult to remove from solution by dialysis, but Tween® 20 can be removed by detergent removing gels.
  • the polyoxyethylene chain found in these detergents makes them subject to oxidation (peroxide formation) as is true with the Triton® X and Brij® series detergents.
  • the zwitterionic detergent is a sulfobetaine derivative of cholic acid. This zwitterionic detergent is useful for membrane protein solubilization when protein activity is important. This detergent is useful over a wide range of pH (pH 2-12) and is easily removed from solution by dialysis due to high CMCs (8-10 mM). This detergent has low absorbances at 280 nm making it useful when protein monitoring at this wavelength is necessary.
  • CHAPS is compatible with the BCA Protein Assay and can be removed from solution by detergent removing gel. Proteins can be iodinated in the presence of CHAPS
  • CHAPS has been successfully used to solubilize intrinsic membrane proteins and receptors and maintain the functional capability of the protein.
  • cytochrome P-450 is solubilized in either Triton® X-100 or sodium cholate aggregates are formed.
  • the French pressure cell employs pressures of 10.4 ⁇ 10 7 Pa (16,000 p.s.i) to break cells open.
  • These apparatus consists of a stainless steel chamber which opens to the outside by means of a needle valve.
  • the cell suspension is placed in the chamber with the needle valve in the closed position.
  • the valve is opened and the piston pushed in to force out any air in the chamber.
  • the chamber is restored to its original position, placed on a solid based and the required pressure is exerted on the piston by a hydraulic press.
  • the needle valve is opened fractionally to slightly release the pressure and as the cells expand they burst.
  • the valve is kept open while the pressure is maintained so that there is a trickle of ruptured cell which may be collected.
  • Mechanical shearing with abrasives may be achieved in Mickle shakers which oscillate suspension vigorously (300-3000 time/min) in the presence of glass beads of 500 mm diameter. This method may result in organelle damage.
  • a more controlled method is to use a Hughes press where a piston forces most cells together with abrasives or deep frozen paste of cells through a 0.25 mm diameter slot in the pressure chamber. Pressures of up to 5.5 ⁇ 10 7 Pa (8000 p.s.i.) may be used to lyse bacterial preparations.
  • blenders which use high speed reciprocating or rotating blades, homogenizers which use an upward/downward motion of a plunger and ball and microfluidizers or impinging jets which use high velocity passage through small diameter tubes or high velocity impingement of two fluid streams.
  • the blades of blenders are inclined at different angles to permit efficient mixing.
  • Homogenizers are usually operated in short high speed bursts of a few seconds to minimize local heat. These techniques are not generally suitable for microbial cells but even very gentle liquid shear is usually adequate to disrupt animal cells.
  • the present invention involve methods of producing an adenovirus that involve isolating the adenovirus.
  • Methods of isolating the adenovirus from host cells include any methods known to those of skill in the art. For example, these methods may include clarification, concentration and diafiltration.
  • One step in the purification process can include clarification of the cell lysate to remove large particulate matter, particularly cellular components, from the cell lysate. Clarification of the lysate can be achieved using a depth filter or by tangential flow filtration.
  • the cell lysate is concentrated. Concentrating the crude cell lysate may include any step known to those of skill in the art.
  • the crude cell lysate may be passed through a depth filter, which consists of a packed column of relatively non-adsorbent material (e.g. polyester resins, sand, diatomeceous earth, colloids, gels, and the like).
  • a depth filter which consists of a packed column of relatively non-adsorbent material (e.g. polyester resins, sand, diatomeceous earth, colloids, gels, and the like).
  • relatively non-adsorbent material e.g. polyester resins, sand, diatomeceous earth, colloids, gels, and the like.
  • TMF tangential flow filtration
  • Membranes are generally arranged within various types of filter apparatus including open channel plate and frame, hollow fibers, and tubules.
  • the resultant virus supernatant may be concentrated and buffer may be exchanged by diafiltration.
  • the virus supernatant can be concentrated by tangential flow filtration across an ultrafiltration membrane of 100-300K nominal molecular weight cutoff.
  • Ultrafiltration is a pressure-modified convective process that uses semi-permeable membranes to separate species by molecular size, shape and/or charge. It separates solvents from solutes of various sizes, independent of solute molecular size. Ultrafiltration is gentle, efficient and can be used to simultaneously concentrate and desalt solutions.
  • Ultrafiltration membranes generally have two distinct layers: a thin (0.1-1.5 ⁇ m), dense skin with a pore diameter of 10-400 angstroms and an open substructure of progressively larger voids which are largely open to the permeate side of the ultrafilter. Any species capable of passing through the pores of the skin can therefore freely pass through the membrane.
  • a membrane is selected that has a nominal molecular weight cut-off well below that of the species being retained. In macromolecular concentration, the membrane enriches the content of the desired biological species and provides filtrate cleared of retained substances. Microsolutes are removed convectively with the solvent. As concentration of the retained solute increases, the ultrafiltration rate diminishes.
  • Some embodiments of the present invention involve use of exchanging buffer of the crude cell lysate.
  • Buffer exchange, or diafiltration involves using ultrafilters is an ideal way for removal and exchange of salts, sugars, non-aqueous solvents separation of free from bound species, removal of material of low molecular weight, or rapid change of ionic and pH environments.
  • Microsolutes are removed most efficiently by adding solvent to the solution being ultrafiltered at a rate equal to the ultrafiltration rate. This washes microspecies from the solution at constant volume, purifying the retained species.
  • Certain embodiments of the methods for producing an adenovirus involve reducing the concentration of contaminating nucleic acids in a crude cell lysate.
  • the present invention employs nucleases to remove contaminating nucleic acids.
  • Exemplary nucleases include Benzonase®, Pulmozyme®; or any other DNase or RNase commonly used within the art.
  • Enzymes such as Benzonaze® degrade nucleic acid and have no proteolytic activity.
  • the ability of Benzonase® to rapidly hydrolyze nucleic acids makes the enzyme ideal for reducing cell lysate viscosity. It is well known that nucleic acids may adhere to cell derived particles such as viruses. The adhesion may interfere with separation due to agglomeration, change in size of the particle or change in particle charge, resulting in little if any product being recovered with a given purification scheme.
  • Benzonase® is well suited for reducing the nucleic acid load during purification, thus eliminating the interference and improving yield.
  • Benzonase® hydrolyzes internal phosphodiester bonds between specific nucleotides. Upon complete digestion, all free nucleic acids present in solution are reduced to oligonucleotides 2 to 4 bases in length.
  • Certain embodiments of the invention involve methods for producing an adenovirus that include purifying the adenovirus.
  • the adenovirus generated in the claimed methods can be purified by any method known to those of skill in the art. In certain embodiments, specific purification techniques are employed. These techniques are delineated as follows.
  • Particle separation by the rate zonal technique is based upon differences in size or sedimentation rates.
  • the technique involves carefully layering a sample solution on top of a performed liquid density gradient, the highest density of which exceeds that of the densest particles to be separated. The sample is then centrifuged until the desired degree of separation is effected, i.e., for sufficient time for the particles to travel through the gradient to form discrete zones or bands which are spaced according to the relative velocities of the particles. Since the technique is time dependent, centrifugation must be terminated before any of the separated zones pellet at the bottom of the tube.
  • the method has been used for the separation of enzymes, hormones, RNA-DNA hybrids, ribosomal subunits, subcellular organelles, for the analysis of size distribution of samples of polysomes and for lipoprotein fractionations.
  • the sample is layered on top of a continuous density gradient which spans the whole range of the particle densities which are to be separated.
  • the maximum density of the gradient therefore, must always exceed the density of the most dense particle.
  • Isopycnic centrifugation in contrast to the rate zonal technique, is an equilibrium method, the particles banding to form zones each at their own characteristic buoyant density.
  • a gradient range can be selected in which unwanted components of the mixture will sediment to the bottom of the centrifuge tube whilst the particles of interest sediment to their respective isopycnic positions.
  • Isopycnic centrifugation depends solely upon the buoyant density of the particle and not its shape or size and is independent of time.
  • the sample is initially mixed with the gradient medium to give a solution of uniform density, the gradient ‘self-forming’, by sedimentation equilibrium, during centrifugation.
  • the equilibrium isodensity method use is generally made of the salts of heavy metals (e.g., caesium or rubidium), sucrose, colloidal silica or Metrizamide.
  • the sample e.g., DNA
  • a concentrated solution of caesium chloride e.g., a concentrated solution of caesium chloride.
  • Centrifugation of the concentrated caesium chloride solution results in the sedimentation of the CsCl molecules to form a concentration gradient and hence a density gradient.
  • the sample molecules (DNA) which were initially uniformly distributed throughout the tube now either rise or sediment until they reach a region where the solution density is equal to their own buoyant density, i.e. their isopycnic position, where they will band to form zones.
  • This technique suffers from the disadvantage that often very long centrifugation times (e.g., 36 to 48 hours) are required to establish equilibrium.
  • adenovirus will be purified using chromatography.
  • the adenovirus may be subjected to one or more chromatography steps.
  • Purification techniques are well known to those of skill in the art. These techniques tend to involve the fractionation of the cellular milieu to separate the adenovirus particles from other components of the mixture. Having separated adenoviral particles from the other components, the adenovirus may be purified using chromatographic and electrophoretic techniques to achieve complete purification.
  • Analytical methods particularly suited to the preparation of a pure adenovrial particle of the present invention are ion-exchange chromatography, size exclusion chromatography; polyacrylamide gel electrophoresis.
  • a particularly efficient purification method to be employed in conjunction with the present invention is HPLC.
  • Certain aspects of the present invention concern the purification, and in particular embodiments, the substantial purification, of an adenoviral particle.
  • the term “purified” as used herein, is intended to refer to a composition, isolatable from other components, wherein the adenoviral particle is purified to any degree relative to its naturally-obtainable form.
  • a purified adenoviral particle therefore also refers to an adenoviral component, free from the environment in which it may naturally occur.
  • purified will refer to an adenoviral particle that has been subjected to fractionation to remove various other components, and which composition substantially retains its expressed biological activity. Where the term “substantially purified” is used, this designation will refer to a composition in which the particle, protein or peptide forms the major component of the composition, such as constituting about 50% or more of the constituents in the composition.
  • Various methods for quantifying the degree of purification of a protein or peptide will be known to those of skill in the art in light of the present disclosure. These include, for example, determining the specific activity of an active fraction, or assessing the amount of polypeptides within a fraction by SDS/PAGE analysis.
  • a preferred method for assessing the purity of a fraction is to calculate the specific activity of the fraction, to compare it to the specific activity of the initial extract, and to thus calculate the degree of purity, herein assessed by a “-fold purification number”.
  • the actual units used to represent the amount of activity will, of course, be dependent upon the particular assay technique chosen to follow the purification and whether or not the expressed protein or peptide exhibits a detectable activity.
  • Partial purification may be accomplished by using fewer purification steps in combination, or by utilizing different forms of the same general purification scheme. For example, it is appreciated that a cation-exchange column chromatography performed utilizing an HPLC apparatus will generally result in a greater-fold purification than the same technique utilizing a low pressure chromatography system. Methods exhibiting a lower degree of relative purification may have advantages in total recovery of protein product, or in maintaining the activity of an expressed protein.
  • chromatographic techniques and other purification techniques known to those of skill in the art may also be employed to purify proteins expressed by the adenoviral vectors of the present invention.
  • Ion exchange chromatography and high performance liquid chromatography are exemplary purification techniques employed in the purification of adenoviral particles and are described in further detail herein below.
  • Ion-Exchange Chromatography The basic principle of ion-exchange chromatography is that the affinity of a substance for the exchanger depends on both the electrical properties of the material and the relative affinity of other charged substances in the solvent. Hence, bound material can be eluted by changing the pH, thus altering the charge of the material, or by adding competing materials, of which salts are but one example. Because different substances have different electrical properties, the conditions for release vary with each bound molecular species. In general, to get good separation, the methods of choice are either continuous ionic strength gradient elution or stepwise elution.
  • An ion exchanger is a solid that has chemically bound charged groups to which ions are electrostatically bound; it can exchange these ions for ions in aqueous solution. Ion exchangers can be used in column chromatography to separate molecules according to charge; actually other features of the molecule are usually important so that the chromatographic behavior is sensitive to the charge density, charge distribution, and the size of the molecule.
  • the principle of ion-exchange chromatography is that charged molecules adsorb to ion exchangers reversibly so that molecules can be bound or eluted by changing the ionic environment. Separation on ion exchangers is usually accomplished in two stages: first, the substances to be separated are bound to the exchanger, using conditions that give stable and tight binding; then the column is eluted with buffers of different pH, ionic strength, or composition and the components of the buffer compete with the bound material for the binding sites.
  • An ion exchanger is usually a three-dimensional network or matrix that contains covalently linked charged groups. If a group is negatively charged, it will exchange positive ions and is a cation exchanger.
  • a typical group used in cation exchangers is the sulfonic group, SO 3 ⁇ . If an H + is bound to the group, the exchanger is said to be in the acid form; it can, for example, exchange one H + for one Na + or two H + for one Ca 2+ .
  • the sulfonic acid group is called a strongly acidic cation exchanger.
  • Other commonly used groups are phenolic hydroxyl and carboxyl, both weakly acidic cation exchangers.
  • If the charged group is positive—for example, a quaternary amino group—it is a strongly basic anion exchanger.
  • the most common weakly basic anion exchangers are aromatic or aliphatic amino groups.
  • the matrix can be made of various material. Commonly used materials are dextran, cellulose, agarose and copolymers of styrene and vinylbenzene in which the divinylbenzene both cross-links the polystyrene strands and contains the charged groups. Table 4 gives the composition of many ion exchangers.
  • the total capacity of an ion exchanger measures its ability to take up exchangeable groups per milligram of dry weight. This number is supplied by the manufacturer and is important because, if the capacity is exceeded, ions will pass through the column without binding.
  • Matrix Exchanger Functional Group Tradename Dextran Strong Cationic Sulfopropyl SP-Sephadex Weak Cationic Carboxymethyl CM-Sephadex Strong Anionic Diethyl-(2- QAE-Sephadex hydroxypropyl)-aminoethyl Weak Anionic Diethylaminoethyl DEAE-Sephadex Cellulose Cationic Carboxymethyl CM-Cellulose Cationic Phospho P-cel Anionic Diethylaminoethyl DEAE-cellulose Anionic Polyethylenimine PEI-Cellulose Anionic Benzoylated- DEAE(BND)-cellulose naphthoylated, deieth
  • the available capacity is the capacity under particular experimental conditions (i.e., pH, ionic strength).
  • pH the extent to which an ion exchanger is charged depends on the pH (the effect of pH is smaller with strong ion exchangers).
  • Another factor is ionic strength because small ions near the charged groups compete with the sample molecule for these groups. This competition is quite effective if the sample is a macromolecule because the higher diffusion coefficient of the small ion means a greater number of encounters.
  • buffer concentration increases, competition becomes keener.
  • the porosity of the matrix is an important feature because the charged groups are both inside and outside the matrix and because the matrix also acts as a molecular sieve. Large molecules may be unable to penetrate the pores; so the capacity will decease with increasing molecular dimensions.
  • the porosity of the polystyrene-based resins is determined by the amount of cross-linking by the divinylbenzene (porosity decreases with increasing amounts of divinylbenzene). With the Dowex and AG series, the percentage of divinylbenzene is indicated by a number after an X—hence, Dowex 50-X8 is 8% divinylbenzene
  • Ion exchangers come in a variety of particle sizes, called mesh size. Finer mesh means an increased surface-to-volume ration and therefore increased capacity and decreased time for exchange to occur for a given volume of the exchanger. On the other hand, fine mesh means a slow flow rate, which can increase diffusional spreading. The use of very fine particles, approximately 10 ⁇ m in diameter and high pressure to maintain an adequate flow is called high-performance or high-pressure liquid chromatography or simply HPLC. Such a collection of exchangers having such different properties—charge, capacity, porosity, mesh—makes the selection of the appropriate one for accomplishing a particular separation difficult.
  • the exchanger is to be anionic or cationic. If the materials to be bound to the column have a single charge (i.e., either plus or minus), the choice is clear. However, many substances (e.g., proteins, viruses), carry both negative and positive charges and the net charge depends on the pH. In such cases, the primary factor is the stability of the substance at various pH values. Most proteins have a pH range of stability (i.e., in which they do not denature) in which they are either positively or negatively charged. Hence, if a protein is stable at pH values above the isoelectric point, an anion exchanger should be used; if stable at values below the isoelectric point, a cation exchanger is required.
  • strong and weak exchangers are also based on the effect of pH on charge and stability. For example, if a weakly ionized substance that requires very low or high pH for ionization is chromatographed, a strong ion exchanger is called for because it functions over the entire pH range. However, if the substance is labile, weak ion exchangers are preferable because strong exchangers are often capable of distorting a molecule so much that the molecule denatures. The pH at which the substance is stable must, of course, be matched to the narrow range of pH in which a particular weak exchanger is charged.
  • Weak ion exchangers are also excellent for the separation of molecules with a high charge from those with a small charge, because the weakly charged ions usually fail to bind. Weak exchangers also show greater resolution of substances if charge differences are very small. If a macromolecule has a very strong charge, it may be impossible to elute from a strong exchanger and a weak exchanger again may be preferable. In general, weak exchangers are more useful than strong exchangers.
  • the Sephadex and Bio-gel exchangers offer a particular advantage for macromolecules that are unstable in low ionic strength. Because the cross-links in these materials maintain the insolubility of the matrix even if the matrix is highly polar, the density of ionizable groups can be made several times greater than is possible with cellulose ion exchangers.
  • the increased charge density means increased affinity so that adsorption can be carried out at higher ionic strengths.
  • these exchangers retain some of their molecular sieving properties so that sometimes molecular weight differences annul the distribution caused by the charge differences; the molecular sieving effect may also enhance the separation.
  • the cellulose ion exchangers have proved to be the best for purifying large molecules such as proteins and polynucleotides. This is because the matrix is fibrous, and hence all functional groups are on the surface and available to even the largest molecules. In many cases however, beaded forms such as DEAE-Sephacel and DEAE-Biogel P are more useful because there is a better flow rate and the molecular sieving effect aids in separation.
  • buffers themselves consist of ions, they can also exchange, and the pH equilibrium can be affected. To avoid these problems, the rule of buffers is adopted: use cationic buffers with anion exchangers and anionic buffers with cation exchangers. Because ionic strength is a factor in binding, a buffer should be chosen that has a high buffering capacity so that its ionic strength need not be too high. Furthermore, for best resolution, it has been generally found that the ionic conditions used to apply the sample to the column (the so-called starting conditions) should be near those used for eluting the column.
  • High Performance Liquid Chromatography is characterized by a very rapid separation with extraordinary resolution of peaks. This is achieved by the use of very fine particles and high pressure to maintain an adequate flow rate. Separation can be accomplished in a matter of minutes, or at most an hour. Moreover, only a very small volume of the sample is needed because the particles are so small and close-packed that the void volume is a very small fraction of the bed volume. Also, the concentration of the sample need not be very great because the bands are so narrow that there is very little dilution of the sample.
  • the methods of producing an adenovirus will involve methods to analyze virus production. Any method known to those of skill in the art can be used to analyze virus production. In a certain embodiment, virus production is analyzed using HPLC.
  • the present invention includes, in certain embodiments, methods for producing an adenovirus that involve placing the adenovirus into a pharmaceutically acceptable composition.
  • the present invention also includes compositions of adenovirus prepared by one of the processes disclosed herein, wherein the composition is a pharmaceutically acceptable composition.
  • composition refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate.
  • pharmaceutically acceptable composition includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the composition.
  • the composition can include supplementary inactive ingredients.
  • the composition for use as a mouthwash may include a flavorant or the composition may contain supplementary ingredients to make the formulation timed-release.
  • Aqueous compositions of the present invention comprise an effective amount of the expression cassette, dissolved or dispersed in a pharmaceutically acceptable carrier or acqueous medium. Such compositions also are referred to as inocula. Examples of aqueous compositions include a formulation for intravenous administration or a formulation for topical application.
  • Solutions of the active compounds as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions also can be prepared in glycerol, liquid polyethylene glycols, mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the adenovirus preparations of the present invention may include classic pharmaceutical preparations. Administration of therapeutic compositions according to the present invention will be via any common route so long as the target tissue is available via that route. Such compositions would normally be administered as pharmaceutically acceptable compositions that include physiologically acceptable carriers, buffers or other excipients.
  • compositions of the present invention are advantageously administered in the form of liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to topical use may also be prepared.
  • a typical composition for such purpose comprises a pharmaceutically acceptable carrier.
  • the composition may contain 10 mg, 25 mg, 50 mg or up to about 100 mg of human serum albumin per ml of phosphate buffered saline.
  • Other pharmaceutically acceptable carriers include aqueous solutions, non-toxic excipients, including salts, preservatives, buffers and the like. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oil and injectable organic esters such as ethyloleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, saline solutions, parenteral vehicles such as sodium chloride, Ringer's dextrose, etc.
  • Preservatives include antimicrobial agents, anti-oxidants, chelating agents and inert gases.
  • the pH and exact concentration of the various components of the pharmaceutical composition are adjusted according to well-known parameters.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and/or the like. These compositions take the form of solutions such as mouthwashes and mouthrinses, suspensions, tablets, pills, capsules, sustained release formulations and/or powders.
  • oral pharmaceutical compositions will comprise an inert diluent and/or assimilable edible carrier, and/or they may be enclosed in hard and/or soft shell gelatin capsule, and/or they may be compressed into tablets, and/or they may be incorporated directly with the food of the diet.
  • the active compounds may be incorporated with excipients and/or used in the form of ingestible tablets, buccal tables, troches, capsules, elixirs, suspensions, syrups, wafers, and/or the like.
  • Such compositions and/or preparations should contain at least 0.1% of active compound.
  • the percentage of the compositions and/or preparations may, of course, be varied and/or may conveniently be between about 2 to about 75% of the weight of the unit, and/or preferably between 25-60%.
  • the amount of active compounds in such therapeutically useful compositions is such that a suitable dosage will be obtained.
  • the tablets, troches, pills, capsules and/or the like may also contain the following: a binder, as gum tragacanth, acacia, cornstarch, and/or gelatin; excipients, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and/or the like; a lubricant, such as magnesium stearate; and/or a sweetening agent, such as sucrose, lactose and/or saccharin may be added and/or a flavoring agent, such as peppermint, oil of wintergreen, and/or cherry flavoring.
  • a binder as gum tragacanth, acacia, cornstarch, and/or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and/or the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as
  • the expression cassette of the present invention may be incorporated with excipients and used in the form of non-ingestible mouthwashes and dentifrices.
  • a mouthwash may be prepared incorporating the active ingredient in the required amount in an appropriate solvent, such as a sodium borate solution (Dobell's Solution).
  • the active ingredient may be incorporated into an antiseptic wash containing sodium borate, glycerin and potassium bicarbonate.
  • the active ingredient also may be dispersed in dentifrices, including: gels, pastes, powders and slurries.
  • the active ingredient may be added in a therapeutically effective amount to a paste dentifrice that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants.
  • compositions of the present invention may be formulated in a neutral or salt form.
  • Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • One example is a spray for administration to the aerodigestive tract.
  • the sprays are isotonic and/or slightly buffered to maintain a pH of 5.5 to 6.5.
  • antimicrobial preservatives similar to those used in ophthalmic preparations, and/or appropriate drug stabilizers, if required, may be included in the formulation.
  • Additional formulations which are suitable for other modes of administration include vaginal or rectal suppositories and/or pessaries.
  • Formulations for other types of administration that is topical include, for example, a cream, suppository, ointment or salve.
  • the adenoviral vector used were Adp53 and Admda7.
  • the cells were HEK293 cells (cell passage #54).
  • the experiments were carried out in 293 cells grown in T-150 flasks (Corning) using an incubator (Form a Scientific, Inc., model # Steri-Cult 200).
  • the infection medium was DMEM+2% FBS.
  • the multiplicity of infection (MOI) was 150 vp/cell.
  • Virus production was reduced significantly at 39° C. relatively to 37° C. (FIG. 1A and FIG. 1B).
  • Admda7 virus yield was 2.6E10 vp/ml or 26,000 vp/cell.
  • Ad-mda7 virus yield was 4.1E10 vp/ml or 41,000 vp/cell.
  • Approximately equivalent virus production was attained at both 37° C. and 35° C.
  • Virus production at 32° C. was much lower than that at 37° C. The same general conclusion was reached for both Adp53 and Admda7 vectors.
  • the optimal infection temperature for adenovirus production is in the range of greater than 32° C. and less than 37° C.
  • the results also demonstrated the detrimental effect of high infection temperature (39° C.) on adenovirus production.
  • Bioreactor model 20/50EH (Wave Biotech, LLC) Culture medium: CD293 protein free medium (Invitrogen TM) HEK293 suspension cells: Adapted to serum-free suspension culture at Introgen Therapeutics, Inc. Adenoviral vectors: Supplied by Introgen Therapeutics, Inc. YSI biochemistry analyzer: YSI-2700 SELECT TM
  • adenoviral vector production run was carried out in a 10 L (5 L working volume) Wave Bioreactor®.
  • HEK293 suspension cells were seeded at 4.8E5 cells/ml and were allowed to grow to 1.2E6 cells/ml in protein-free CD293 medium.
  • the rocking speed was set at 10 and the rocking angle was set at 11.
  • the culture pH was maintained by adjusting CO 2 gas percentage delivered by the gas mixer.
  • the dissolved oxygen tension (DOT) in the culture medium was monitored using a disposable DOT probe supplied by Wave BiotechTM.
  • FIG. 2 demonstrates that cell concentration peaked at day 4, and viability remained steady but showed decline after day 5.
  • FIG. 3 shows nutrient/metabolite data. Following virus infection, glucose concentration declined over time, and lactate concentration increased over time.
  • Adenoviral vector production was measured using an anion exchange HPLC method.
  • the adenoviral vector concentration in the bioreactor was found to be 4.5E10 vp/ml, and the cell-specific vector productivity was 37,000 vp/cell.
  • the vector productivity is in close agreement with those seen using the parental anchorage dependent HEK293 cells in serum-containing media.
  • FIG. 4 A uniquely designed perfusion Wave bioreactor was set up as shown in FIG. 4.
  • the system shown in FIG. 4 utilizes an internal flat filter to provide separation between the cells and spent medium. Spent culture medium is withdrawn through the floating filter. No medium recirculation is required, and consequently this mode of medium perfusion is very gentle to the cells in culture. The wave action minimizes filter clogging during perfusion. The culture volume during perfusion is maintained by a load cell used to trigger fresh medium addition.
  • a 1 L (working volume) perfusion Wave bioreactor was used to increase the cell concentration in the growth phase prior to virus infection.
  • the cell growth and nutrient/metabolite concentrations during culture are shown in FIG. 5 and FIG. 6.
  • HEK293 suspension cells were seeded at a cell concentration of 5E5 cells/ml. On day 3 of culture, medium perfusion was started at a cell concentration of 1.7E6 cells/ml. Cell concentration increased approximately exponentially to. 1E7 cells/ml on day 6, and cell viability was maintained above 90%.
  • the culture was diluted 10-fold with fresh CD293 medium to supplement nutrients and dilute potentially toxic metabolites.
  • the culture was immediately infected with an adenoviral vector at a MOI of 50 vp/cell in a larger Wave Bioreactor® without a perfusion filter. Medium perfusion was not resumed after virus infection. The infection was allowed to proceed for 4 days. The culture was harvested on day 4 post-virus infection.
  • the adenoviral vector production was measured by an anion exchange HPLC method.
  • the adenoviral vector concentration in the Wave Bioreactor® was 3.5E10 vp/ml, and the cell-specific vector productivity was 35,000 vp/cell.
  • Wilson Nature, 365:691-692, 1993.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biophysics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Plant Pathology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Endocrinology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US10/439,278 2003-05-15 2003-05-15 Methods and compositions for the production of adenoviral vectors Abandoned US20040229335A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
US10/439,278 US20040229335A1 (en) 2003-05-15 2003-05-15 Methods and compositions for the production of adenoviral vectors
KR1020057021606A KR20060028388A (ko) 2003-05-15 2004-05-14 아데노바이러스 벡터의 생산을 위한 방법과 조성물
PCT/US2004/015009 WO2004104190A2 (en) 2003-05-15 2004-05-14 Methods and compositions for the production of adenoviral vectors
CNA2004800185873A CN1816620A (zh) 2003-05-15 2004-05-14 用于产生腺病毒载体的方法和组合物
CA002525603A CA2525603A1 (en) 2003-05-15 2004-05-14 Methods and compositions for the production of adenoviral vectors
JP2006533032A JP2007500015A (ja) 2003-05-15 2004-05-14 アデノウイルスベクター産生のための方法および組成物
EP04752113A EP1627057A2 (en) 2003-05-15 2004-05-14 Methods and compositions for the production of adenoviral vectors
AU2004241421A AU2004241421A1 (en) 2003-05-15 2004-05-14 Methods and compositions for the production of adenoviral vectors
US11/079,986 US7419808B2 (en) 2003-05-15 2005-03-15 Methods and compositions for the production of adenoviral vectors
IL171904A IL171904A0 (en) 2003-05-15 2005-11-10 Methods and compositions for the production of adenoviral vectors

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10/439,278 US20040229335A1 (en) 2003-05-15 2003-05-15 Methods and compositions for the production of adenoviral vectors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/079,986 Division US7419808B2 (en) 2003-05-15 2005-03-15 Methods and compositions for the production of adenoviral vectors

Publications (1)

Publication Number Publication Date
US20040229335A1 true US20040229335A1 (en) 2004-11-18

Family

ID=33417767

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/439,278 Abandoned US20040229335A1 (en) 2003-05-15 2003-05-15 Methods and compositions for the production of adenoviral vectors
US11/079,986 Expired - Lifetime US7419808B2 (en) 2003-05-15 2005-03-15 Methods and compositions for the production of adenoviral vectors

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/079,986 Expired - Lifetime US7419808B2 (en) 2003-05-15 2005-03-15 Methods and compositions for the production of adenoviral vectors

Country Status (9)

Country Link
US (2) US20040229335A1 (ko)
EP (1) EP1627057A2 (ko)
JP (1) JP2007500015A (ko)
KR (1) KR20060028388A (ko)
CN (1) CN1816620A (ko)
AU (1) AU2004241421A1 (ko)
CA (1) CA2525603A1 (ko)
IL (1) IL171904A0 (ko)
WO (1) WO2004104190A2 (ko)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020177215A1 (en) * 1996-11-20 2002-11-28 Introgen Therapeutics, Inc. Methods for producing purified adenoviral vectors
US20050158283A1 (en) * 2003-05-15 2005-07-21 Shuyuan Zhang Methods and compositions for the production of adenoviral vectors
US20050226794A1 (en) * 2004-02-03 2005-10-13 Geoffrey Hodge System and method for manufacturing
US20050272146A1 (en) * 2004-06-04 2005-12-08 Geoffrey Hodge Disposable bioreactor systems and methods
US20080139865A1 (en) * 2006-07-14 2008-06-12 Xcellerex, Inc. Environmental containment systems
US20090135667A1 (en) * 2004-01-07 2009-05-28 Terentiev Alexandre N Mixing bag with integral sparger and sensor receiver
US20090253184A1 (en) * 2008-01-23 2009-10-08 Introgen Therapeutics, Inc. Compositions and methods related to an adenoviral trans-complementing cell line
CN101799459A (zh) * 2010-03-31 2010-08-11 天水师范学院 一种测定多糖磷酸酯中磷含量的方法
CN102219848A (zh) * 2011-05-25 2011-10-19 浙江海正药业股份有限公司 重组人干扰素β-1a的纯化方法
WO2015120527A3 (en) * 2014-02-17 2015-12-10 Advantech Bioscience Farmacêutica Ltda Enhancement of recombinant protein expression using a membrane-based cell retention system
US9339026B2 (en) 2012-06-14 2016-05-17 Therapeutic Proteins International, LLC Pneumatically agitated and aerated single-use bioreactor
US9855319B2 (en) 2014-04-01 2018-01-02 Advantech Bioscience Farmacêutica Ltda Stabilization of factor VIII without calcium as an excipient
US9907835B2 (en) 2014-04-01 2018-03-06 Advanced Bioscience Farmacêutica LTDA Stable Factor VIII formulations with low sugar-glycine
CN110225756A (zh) * 2016-12-02 2019-09-10 鲁比厄斯治疗法股份有限公司 与用于穿透实体瘤的细胞系统相关的组合物和方法
EP3256574B1 (en) 2015-02-09 2020-11-25 INSERM - Institut National de la Santé et de la Recherche Médicale Recombinant adeno-associated virus particle purification comprising an affinity purification step
CN114591414A (zh) * 2022-03-24 2022-06-07 江西浩然生物制药有限公司 一种人绒毛膜促性腺激素的制备方法

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2884255B1 (fr) * 2005-04-11 2010-11-05 Vivalis Utilisation de lignees de cellules souches aviaires ebx pour la production de vaccin contre la grippe
ITRM20050191A1 (it) * 2005-04-20 2006-10-21 Consiglio Nazionale Ricerche Vettore adenovirale ricombinante di espressione di proteine della famiglia mdm e usi relativi.
WO2007059473A2 (en) * 2005-11-12 2007-05-24 Introgen Therapeutics, Inc. Methods for the production and purification of adenoviral vectors
EP1966842B1 (en) 2005-11-30 2019-08-28 Lomox Limited Lighting elements and devices
EP1808697A1 (en) * 2006-01-13 2007-07-18 Novartis Vaccines and Diagnostics, Inc. Use of an ion exchange matrix for determining the concentration of virus particles and/or virus antigens
US8785193B2 (en) * 2006-09-14 2014-07-22 The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Dissection tool and methods of use
EP1942192A1 (en) * 2007-01-08 2008-07-09 Heinrich-Pette-Institut für experimentelle Virologie und Immunologie Use of a tailored recombinase for the treatment of retroviral infections
US9109193B2 (en) * 2007-07-30 2015-08-18 Ge Healthcare Bio-Sciences Corp. Continuous perfusion bioreactor system
DE102008015386B4 (de) * 2008-03-20 2015-10-01 Sartorius Stedim Biotech Gmbh Bioreaktor
US10273462B2 (en) * 2011-07-27 2019-04-30 Vectalys Sas Virus-based vector compositions useful for transducing eukaryotic cells
CN102552938A (zh) * 2012-03-22 2012-07-11 南京大学 Egr-1拮抗剂在制备治疗Ⅱ型糖尿病的药物中的用途
CN103602625B (zh) * 2013-08-16 2015-09-30 苏州大学 含有重组腺病毒质粒的大肠杆菌及重组腺病毒的应用
CN103881984A (zh) * 2013-12-06 2014-06-25 深圳市赛百诺基因技术有限公司 无血清悬浮细胞生产重组腺病毒的方法及其药品制剂生产方法
KR20170016386A (ko) * 2014-06-02 2017-02-13 테티스, 아이엔씨. 개질된 생체중합체, 및 이의 제조 및 사용 방법
US9663766B2 (en) * 2015-07-24 2017-05-30 Bio-Rad Laboratories, Inc. Methods for purifying adenovirus vectors
CA3004346A1 (en) 2015-11-23 2017-06-01 Tethis, Inc. Coated particles and methods of making and using the same
CN105647852A (zh) * 2016-03-30 2016-06-08 北京普瑞金科技有限公司 用于慢病毒包装的无血清培养293t细胞的方法
CN106978408B (zh) * 2017-03-29 2020-06-16 武汉新华扬生物股份有限公司 一种食品级中温α-淀粉酶的生产方法
CN107384873A (zh) * 2017-09-05 2017-11-24 成都汇宇生物技术有限公司 重组腺病毒的纯化方法
CN107723279B (zh) * 2017-10-18 2020-11-10 成都远睿生物技术有限公司 一种缺陷型腺病毒AdC68-GP的培养方法
CN108795885A (zh) * 2018-06-28 2018-11-13 赛诺(深圳)生物医药研究有限公司 生产重组腺病毒的方法
CN111304176A (zh) * 2020-03-07 2020-06-19 北京工业大学 基于q-6xl和4ff的腺病毒载体规模化纯化方法
CN111793611A (zh) * 2020-07-14 2020-10-20 安徽安科生物工程(集团)股份有限公司 一种用生物反应器罐流培养溶瘤腺病毒的工艺优化方法
CN112877358B (zh) * 2021-02-07 2023-02-03 上海市第一人民医院 一种基于5型腺病毒序列的重组质粒及应用
US11299700B1 (en) 2021-02-19 2022-04-12 Acequia Biotechnology, Llc Bioreactor containers and methods of growing hairy roots using the same
WO2024075819A1 (ja) * 2022-10-07 2024-04-11 Agc株式会社 細胞を用いたウイルスベクターの生産方法

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3962424A (en) * 1974-01-31 1976-06-08 Recherche Et Industrie Therapeutiques (R.I.T.) Live brovine adenovirus vaccines, preparation thereof and method of vaccination using them
US4352883A (en) * 1979-03-28 1982-10-05 Damon Corporation Encapsulation of biological material
US5585362A (en) * 1989-08-22 1996-12-17 The Regents Of The University Of Michigan Adenovirus vectors for gene therapy
US5670488A (en) * 1992-12-03 1997-09-23 Genzyme Corporation Adenovirus vector for gene therapy
US5824544A (en) * 1995-03-24 1998-10-20 Genzyme Corporation Adenovirus vectors for gene therapy
US5932210A (en) * 1993-10-25 1999-08-03 Canji Inc. Recombinant adenoviral vector and methods of use
US6194191B1 (en) * 1996-11-20 2001-02-27 Introgen Therapeutics, Inc. Method for the production and purification of adenoviral vectors
US6355622B1 (en) * 1996-08-16 2002-03-12 The Trustees Of Columbia University In The City Of New York Use of a melanoma differentiation associated gene (mda-7) for inducing apoptosis of a tumor cell
US20020182723A1 (en) * 1998-12-01 2002-12-05 Introgen Therapeutics, Inc. An improved method for the production and purification of adenoviral vectors

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0682697A4 (en) 1993-01-29 1997-07-30 New Brunswick Scientific Co METHOD AND APPARATUS FOR CULTURING ANCHOR AND SUSPENSION CELLS.
IL127692A0 (en) * 1996-07-01 1999-10-28 Rhone Poulenc Rorer Sa Method for producing recombinant adenovirus
US6190913B1 (en) * 1997-08-12 2001-02-20 Vijay Singh Method for culturing cells using wave-induced agitation
US6168941B1 (en) * 2000-04-07 2001-01-02 Genvec, Inc. Method of producing adenoviral vector stocks
US6544788B2 (en) * 2001-02-15 2003-04-08 Vijay Singh Disposable perfusion bioreactor for cell culture
AU2003268210A1 (en) * 2002-08-28 2004-03-19 Introgen Therapeutics Inc. Chromatographic methods for adenovirus purification
US20040229335A1 (en) * 2003-05-15 2004-11-18 Introgen Therapeutics, Inc. Methods and compositions for the production of adenoviral vectors
CA2586107A1 (en) * 2004-11-03 2006-05-18 Introgen Therapeutics Inc. A novel method for the production and purification of adenoviral vectors

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3962424A (en) * 1974-01-31 1976-06-08 Recherche Et Industrie Therapeutiques (R.I.T.) Live brovine adenovirus vaccines, preparation thereof and method of vaccination using them
US4352883A (en) * 1979-03-28 1982-10-05 Damon Corporation Encapsulation of biological material
US5585362A (en) * 1989-08-22 1996-12-17 The Regents Of The University Of Michigan Adenovirus vectors for gene therapy
US5670488A (en) * 1992-12-03 1997-09-23 Genzyme Corporation Adenovirus vector for gene therapy
US5932210A (en) * 1993-10-25 1999-08-03 Canji Inc. Recombinant adenoviral vector and methods of use
US5824544A (en) * 1995-03-24 1998-10-20 Genzyme Corporation Adenovirus vectors for gene therapy
US6355622B1 (en) * 1996-08-16 2002-03-12 The Trustees Of Columbia University In The City Of New York Use of a melanoma differentiation associated gene (mda-7) for inducing apoptosis of a tumor cell
US6194191B1 (en) * 1996-11-20 2001-02-27 Introgen Therapeutics, Inc. Method for the production and purification of adenoviral vectors
US20020182723A1 (en) * 1998-12-01 2002-12-05 Introgen Therapeutics, Inc. An improved method for the production and purification of adenoviral vectors

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030008375A1 (en) * 1996-11-20 2003-01-09 Introgen Therapeutics, Inc. Methods for treating patients with adenoviral vectors
US20050089999A1 (en) * 1996-11-20 2005-04-28 Introgen Therapeutics, Inc. Method for the production and purification of adenoviral vectors
US20020177215A1 (en) * 1996-11-20 2002-11-28 Introgen Therapeutics, Inc. Methods for producing purified adenoviral vectors
US7445930B2 (en) * 1996-11-20 2008-11-04 Introgen Therapeutics Inc. Method for the production and purification of adenoviral vectors
US7510875B2 (en) 1996-11-20 2009-03-31 Introgen Therapuetics, Inc. Methods for producing purified adenoviral vectors
US20050158283A1 (en) * 2003-05-15 2005-07-21 Shuyuan Zhang Methods and compositions for the production of adenoviral vectors
US7419808B2 (en) * 2003-05-15 2008-09-02 Introgen Therapeutics, Inc. Methods and compositions for the production of adenoviral vectors
US20100197003A1 (en) * 2004-01-07 2010-08-05 Terentiev Alexandre N Bioreactor
US8123199B2 (en) 2004-01-07 2012-02-28 Atmi Packaging, Inc. Bioreactor
US7992846B2 (en) 2004-01-07 2011-08-09 Atmi Packaging, Inc. Mixing bag with integral sparger and sensor receiver
US20090135667A1 (en) * 2004-01-07 2009-05-28 Terentiev Alexandre N Mixing bag with integral sparger and sensor receiver
US20050226794A1 (en) * 2004-02-03 2005-10-13 Geoffrey Hodge System and method for manufacturing
US9671798B2 (en) 2004-02-03 2017-06-06 Ge Healthcare Bio-Sciences Corp. System and method for manufacturing
US8298054B2 (en) 2004-02-03 2012-10-30 Xcellerex, Inc. System and method for manufacturing
US7629167B2 (en) 2004-06-04 2009-12-08 Xcellerex, Inc. Disposable bioreactor systems and methods
US20050272146A1 (en) * 2004-06-04 2005-12-08 Geoffrey Hodge Disposable bioreactor systems and methods
US20080139865A1 (en) * 2006-07-14 2008-06-12 Xcellerex, Inc. Environmental containment systems
US7819934B2 (en) 2006-07-14 2010-10-26 Xcellerex, Inc. Environmental containment systems
US20090253184A1 (en) * 2008-01-23 2009-10-08 Introgen Therapeutics, Inc. Compositions and methods related to an adenoviral trans-complementing cell line
CN101799459A (zh) * 2010-03-31 2010-08-11 天水师范学院 一种测定多糖磷酸酯中磷含量的方法
CN102219848A (zh) * 2011-05-25 2011-10-19 浙江海正药业股份有限公司 重组人干扰素β-1a的纯化方法
US9339026B2 (en) 2012-06-14 2016-05-17 Therapeutic Proteins International, LLC Pneumatically agitated and aerated single-use bioreactor
WO2015120527A3 (en) * 2014-02-17 2015-12-10 Advantech Bioscience Farmacêutica Ltda Enhancement of recombinant protein expression using a membrane-based cell retention system
US9855319B2 (en) 2014-04-01 2018-01-02 Advantech Bioscience Farmacêutica Ltda Stabilization of factor VIII without calcium as an excipient
US9907835B2 (en) 2014-04-01 2018-03-06 Advanced Bioscience Farmacêutica LTDA Stable Factor VIII formulations with low sugar-glycine
EP3256574B1 (en) 2015-02-09 2020-11-25 INSERM - Institut National de la Santé et de la Recherche Médicale Recombinant adeno-associated virus particle purification comprising an affinity purification step
CN110225756A (zh) * 2016-12-02 2019-09-10 鲁比厄斯治疗法股份有限公司 与用于穿透实体瘤的细胞系统相关的组合物和方法
CN114591414A (zh) * 2022-03-24 2022-06-07 江西浩然生物制药有限公司 一种人绒毛膜促性腺激素的制备方法

Also Published As

Publication number Publication date
WO2004104190A2 (en) 2004-12-02
KR20060028388A (ko) 2006-03-29
AU2004241421A1 (en) 2004-12-02
EP1627057A2 (en) 2006-02-22
IL171904A0 (en) 2006-04-10
JP2007500015A (ja) 2007-01-11
CA2525603A1 (en) 2004-12-02
US7419808B2 (en) 2008-09-02
CN1816620A (zh) 2006-08-09
US20050158283A1 (en) 2005-07-21
WO2004104190A3 (en) 2005-05-19

Similar Documents

Publication Publication Date Title
US7419808B2 (en) Methods and compositions for the production of adenoviral vectors
US9428768B2 (en) Method for the production and purification of adenoviral vectors
US20070172846A1 (en) Methods for the Production and Purification of Adenoviral Vectors
US7445930B2 (en) Method for the production and purification of adenoviral vectors
US9228205B2 (en) Complementing cell lines
US20040106184A1 (en) Chromatographic methods for adenovirus purification
US20060166364A1 (en) Use of flexible bag containers for viral production
WO1998022588A9 (en) An improved method for the production and purification of adenoviral vectors
US20080299545A1 (en) Chromatographic methods for assessing adenovirus purity
US20080044378A1 (en) Methods and Compositions for Protein Production Using Adenoviral Vectors

Legal Events

Date Code Title Description
AS Assignment

Owner name: INTROGEN THERAPEUTICS, INC., TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZHANG, SHUYUAN;PHAM, HAI;REEL/FRAME:014535/0540

Effective date: 20030624

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: CRUCELL HOLLAND B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:INTROGEN THERAPEUTICS, INC.;REEL/FRAME:023279/0858

Effective date: 20090515

Owner name: CRUCELL HOLLAND B.V.,NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:INTROGEN THERAPEUTICS, INC.;REEL/FRAME:023279/0858

Effective date: 20090515