EP0973866A1 - Adenovirus e1-complementing cell lines - Google Patents

Adenovirus e1-complementing cell lines

Info

Publication number
EP0973866A1
EP0973866A1 EP98908661A EP98908661A EP0973866A1 EP 0973866 A1 EP0973866 A1 EP 0973866A1 EP 98908661 A EP98908661 A EP 98908661A EP 98908661 A EP98908661 A EP 98908661A EP 0973866 A1 EP0973866 A1 EP 0973866A1
Authority
EP
European Patent Office
Prior art keywords
deleted
adenovirus
cell
recombinant
mammalian cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP98908661A
Other languages
German (de)
French (fr)
Other versions
EP0973866A4 (en
Inventor
David Ayares
Ramon Alemany
Wei-Wei Zhang
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.)
Genstar Therapeutics Corp
Original Assignee
Baxter International 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 Baxter International Inc filed Critical Baxter International Inc
Publication of EP0973866A1 publication Critical patent/EP0973866A1/en
Publication of EP0973866A4 publication Critical patent/EP0973866A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10345Special targeting system for viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N2710/10352Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • This invention relates to novel cells and methods for use in propagating El- deleted adeno viruses.
  • adenoviral vectors used in gene therapy applications were designed to have deletions in the El region of the adenovirus 5 (Ad5) genome.
  • the El region, not including region IX, consists of 9% of the left end of Ad5 (1.2 - 9.8 map units), and is subdivided into two regions, E1A and E1B, each one coding for several proteins.
  • E1A/E1B is required for virus replication and for expression of all other Ad5 proteins (E2-E4, Late Proteins; Ginsberg, H.S. The Adeno viruses. Plenum Press, New York. p.46-67(1984).
  • El deficient adenoviral vectors are propagated in an Ad5 helper cell line called 293 (Graham, F.L. and Smiley, J, J. Gen. Virol. 36, p.54-72 (1977).
  • 293 cells were derived by transforming human embryonic kidney cells with sheared fragments of Ad5 DNA. Genomic analysis revealed that 293 cells contain four to five copies per cell of the left 12% of the viral genome (including the entire El region) and approximately one copy per cell of 9% of the right end, the E4 region (Aiello,L.,et al, Virology 94, p.460-469 (1979).
  • RCA in 293 cells can present severe ramifications for the safety of human gene therapy trials (Lochmuller, H., et al., Human Gene Therapy 5 . , p. 1485-1491 (1994).
  • recombination in 293 cells can also cause deletions and rearrangements that affect transgene expression, thereby decreasing the titer of functional adenovirus particles.
  • This invention encompasses a series of helper cell lines for the complementation, amplification, and controlled attenuation of El-deleted adenovirus. These cell lines are advantageous because they can complement adenovirus El gene deletions without production of replication competent adenovirus (RCA).
  • a preferred embodiment is an A549E1 cell line that contains only the Ad5 El DNA sequences sufficient for complementation of El -deleted adenoviral vectors without sequences that overlap with the adenovirus vector.
  • the El DNA sequences comprise El A and E1B genes.
  • the present invention embodies methods for selectively propagating mini-adenovirus without generating RCA, by transfecting an A549E1 cell line with DNA sequences that encode a polypeptide sufficient for packaging attenuation of El -deleted helper virus.
  • the polypeptide comprises Cre recombinase.
  • the polypeptide comprises TetR-KRAB.
  • Figure 1 is a diagram indicating the structure of adenovirus sequences in a typical El- deleted Ad helper virus (top line), in 293 cells (ref.5), and in other El -containing cell lines, including 911 cells (ref.8) and A549E1-68 cells (this invention); and how recombination between homologous adenovirus sequences occurs to generate a replication-competent adenovirus (RCA).
  • Figure 2 is a diagram of the CMV-E1 mammalian expression vector.
  • Figure 3 is a Southern blot analysis of G418 r A549E1 clones.
  • Figure 4A is a Western blot analysis of El A protein expression in A549 cells, 293 Cells, and A549El-68.
  • Figure 4B is the metabolic 35 S labeling and immunoprecipitation of EIB proteins in
  • Figure 5 is a representation of the El -deleted adenovirus vector, Ad5-CA-GFP.
  • Figure 6 is an agarose gel analysis of 40 PCR reactions using ElA-specific primers for detection of RCA.
  • Figure 7 is a diagram of a system for the attenuation of helper virus with a loxP -modified packaging signal.
  • Figure 8 is a diagram of the pCMV-Cre-Puro vector.
  • Figure 9 is a diagram of the pBS/loxP-stop/MCLpA vector.
  • Figure 10 is a bar graph depicting luciferase expression in both control cells (A549E1-
  • This invention provides cell lines that can complement El -deleted adenovirus without the disadvantage of undesirable recombination and RCA. These cell lines are obtained by cloning and expressing in the A549 cell line only those sequences that are required for El complementation and excluding from the cell line all other Ad5 sequences that have homology to the vector and could cause recombination to produce RCA.
  • FIG. 1 shows the structure of adenovirus sequences in 293 cells versus other El -containing cell lines (including A549E1 cells), and how recombination between homologous adenovirus sequences occurs to generate a replication-competent adenovirus (RCA).
  • RCA replication-competent adenovirus
  • A549E1-68 contains no sequence overlap with sequences present in the El -deleted Ad helper virus and thus, recombination to produce RCA is not possible (FIG. 1). Characterization of this A549E1 cell line demonstrated the production of El A and EIB proteins, high infectivity with adenovirus vectors, complementation of El- deleted adenovirus to produce high-titer virus stocks, as well as, the lack of production of replication-competent adenovirus (RCA).
  • A549-E1 complementing cell lines which, in addition to producing El, also produce proteins required for further manipulation of adenoviral vectors, thus providing a novel series of RCA-free adenovirus helper cell lines, as tools for novel virus production.
  • the first example is an A549E1 cell line which expresses the Cre recombinase.
  • This invention provides a novel El -deleted helper virus whose packaging signal is flanked by loxP sites, and when this helper virus is propagated in an A549E1 cell line expressing Cre, the packaging signal is deleted by excision, thus attenuating helper virus packaging and enriching for packaging of mini-adenovirus (Ad5 virus which is devoid of all viral protein-coding sequences).
  • This is advantageous because during the production of helper-dependent, mini-adenovirus it becomes necessary to attenuate helper virus packaging in order to enrich for the mini-virus.
  • 293-Cre cells have been generated for this purpose (Parks, R. , et al, P.N.A.S. 93, p.13565-13570 (1996), however, A549E1- Cre cells have an advantage in that they would perform this task in an RCA-free environment.
  • a further embodiment of the present invention includes an A549-E1 complementing cell line which expresses the TetR-KRAB fusion protein, which would be used to amplify, and control the packaging efficiency of an El -deleted helper virus whose packaging signal has been modified to contain multiple tetracycline operator (tetO) sites.
  • tetO tetracycline operator
  • Packaging of the helper virus can be restored by growing the cells and virus in the presence of tetracycline, which binds to the tet-KRAB repressor causing its dissociation from the tetO/packaging signal and a reversal of packaging repression.
  • tetracycline which binds to the tet-KRAB repressor causing its dissociation from the tetO/packaging signal and a reversal of packaging repression.
  • Example 1 Construction of the E1A/E1B vector To generate an expression vector which harbors only the E1A/E1B sequences required for complementation, a 3.1 kb DNA fragment coding for Ad5 El A and EIB genes was cloned in two pieces, sequentially, into the superlinker vector, pSL301 (Invitrogen). First, an 881 bp Afl III to Xbal fragment (Ad5 base pairs 462-1343) was cloned from pBRXad5KpnICl (a subclone of pJM17) into pSL301 (Afl III/XbaI).
  • a contiguous 2194 bp Xbal to Afl II (Ad5 base pairs 1343-3537) was cloned from pBRXad5XhoIClinto the same vector.
  • the resultant 3075 bp El fragment (in pSL301) contains the TATA box and RNA cap site for El A, El A coding sequence, complete EIB promoter, and EIB coding sequence, including the stop codon for EIB p55 protein, but not including region IX.
  • This CMV-El expression plasmid (FIG. 2), was transfected using Lipofectamine (Gibco BRL) into A549 human lung carcinoma cells (ATCC CRL 185) and G418 R colonies were isolated. Single-cell clones were screened for functional E1A/E1B expression.
  • An El -deleted adenovirus containing a green florescence protein (GFP) expression cassette, Ad5 CA-GFP was used to infect the A549-E1 clones. Three days post-infection, clones were screened for production of El -complemented Ad5 CA-GFP adenovirus by visual examination for cytopathic effect (CPE).
  • CPE cytopathic effect
  • A549E1-68 displayed 100% CPE in 3 days, similar to that observed for 293 cells.
  • the clear area in the center of the plaque is evidence of CPE caused by El -complemented virus amplification.
  • This clone also showed high infectivity, in that virtually 100% of the cells fluoresced green 24 hours post-infection.
  • the high infection rate and rapid generation of CPE induced in this cell line is strong evidence that functional E1A/E1B proteins are being produced that are capable of promoting replication and amplification of the El- deleted Ad5-CA-GFP virus.
  • the A459E1 cell was deposited at the American Type Tissue Culture Collection (ATCC) under the Budapest Treaty on January 15, 1998 as ATCC Designation CRL-12458 (viability confirmed January 20, 1998).
  • FIG. 3 shows a Southern blot using an El sequence-specific DNA probe. This assay demonstrated the presence of the CMV-El transgene in A549E1-68 (Lane 4), and a subclone of A549E1-68 (El-68.3), but not in the parental A549 cell line (Lane 2). Sequences hybridizing with the El -specific probe were also observed in 293 cells as expected since they complement El-deleted adenovirus (Lane 3). The morphology of the El-transfected cells was significantly different from the parental A549 cell line.
  • A549 cells at sub-confluent density grow as distinct single cells with an elongated, fibroblast- like morphology, whereas the El cell line A549E1-68 grows as colonies of cells with a more cuboidal morphology.
  • A549E1-68 was also compared with 293 cells for production of El -deleted adenovirus (Ad5 CA-GFP) by plaque assay and found to produce an equivalent titer of complemented virus (7 x 10 9 pfu for A549E1-68 vs. 9 x 10 9 pfu for 293).
  • FIG. 4A shows a Western blot analysis using an EIA specific antibody (M73, Oncogene Science). This antibody detected two ElA-specific bands with apparent molecular weights of 46kd and 42kd in the A549E1-68 cell line (lane 3), corresponding to products expected from EIA 13S and 12S mRNAs (Ginsberg, 1984), and identical in size to those observed in 293 cells (lane 2). These El A-specific bands were not detected in parental A549 cells (lane 1).
  • FIG. 4B shows the immunoprecipitation of metabolically-radiolabeled proteins by a monoclonal antibody specific for EIB p55.
  • A549E1-68 produced an immunoreactive band of approximately 55 kd (lane 3) that was not detected in parental A549 cells. This 55 kd, ElB-specific band, as well as secondary background bands, were observed in 293 cells also (lane 2). Extra "background" bands found in both experimental and control lanes have been observed by other authors and are attributed to co-immunoprecipitation of a variety of proteins including, cyclins, p53, and Rb. It is clear that A549E1-68 not only expresses EIA and EIB, but that they are functional, since this cell line can complement for production of high titer, El -deleted, recombinant adenovirus.
  • Example 3 El-deleted adenovirus produced in A549E1 cells is RCA-free
  • Ad5 helper cell line can complement without production of RCA
  • a series of PCR RCA assays were performed following amplification in A549E1 cells of the E 1 -deleted Ad5 -CA-GFP adenovirus vector.
  • the Ad5-CA-GFP vector is illustrated in FIG. 5. It contains a transcriptional control element consisting of the CMV
  • Ad5-CA-GFP virus was serially propagated through 20 passages on A549E1-68 cells. Following serial propagation and virus amplification, Ad5-CA-GFP virus DNA was isolated by freeze-thaw lysis, and PCR was performed using primers specific for either the EIA region or the E2B region. Amplification of an 880 bp E2B product serves as a PCR positive control, while the presence of a 1086 bp El A-specific product is evidence that an El (+) replication-competent adenovirus (RCA) has been produced during amplification of the El (-) Ad5 -CA-GFP.
  • RCA replication-competent adenovirus
  • Ad5-CA- GFP virus DNA (equivalent to 1 x 10 10 virus particles), obtained from amplification in A549E1 cells, was divided into 40 PCR reactions and tested for RCA using the EIA primers (FIG. 6). For both top and bottom panels of FIG.
  • lane 1 contains 1 kb DNA markers
  • lane 2 contains wild type Ad5 virus DNA
  • lane 3 consists of PCR of Ad5-CA- GFP virus DNA (E1-) isolated from 20+ passages on A549E1-68 cells using EIA and E2B specific primers (positive control)
  • lanes 4-20 consist of PCR of Ad5-CA-GFP virus DNA (E1-) isolated from 20+ passages on A549E1-68 cells, using ElA-specific primers only. No 1086 bp El region specific PCR fragments were detected in any of the reactions indicating that no RCA was present in the virus prep.
  • a second, CPE-based RCA assay was performed by amplifying El -deleted adenovirus (Ad5-CA-GFP) on A549E1-68 cells and testing the amplified virus by passaging, on normal A549 cells (don't make El) for production of El -containing RCA. Plaque formation (CPE) on a monolayer of normal A549 cells would provide evidence for the production of wild-type (El +) virus during amplification on the El helper cell line, A549E1-68. 2 x 10 10 10 El (-) virus particles (amplified using A549E1-68) were used to infect each of five 150mm plates of normal A549 cells (1 x 10" particles total).
  • a 2nd generation El -complementing cell line was generated using the A549E1- 68.3 clonal line for transfection with Cre recombinase.
  • This cell line will both complement El -deleted adenovirus vectors and mediate the excision of sequences surrounded by loxP sites.
  • Our primary use for this cell line is to further attenuate packaging of an Ad5 helper virus, whose packaging signal is flanked by two loxP sites (FIG. 7), in order to enrich for packaging of the desired El -deficient, mini-adenovirus vector.
  • 293 cells expressing the Cre recombinase were generated for a similar purpose by Parks et al. (P.N.A.S.
  • the A549E1-Cre cell line described in this invention will not only attenuate helper virus packaging in a similar fashion, it also has the advantage that any adenovirus produced will be free of deleterious RCA.
  • a Cre expression vector was constructed as a first step towards the production of the A549E1-Cre cell line.
  • a 1440 bp SV40 promo ter-puromycin cassette (for selection in Neo R A549E1 cells) was cloned into a unique EcoRI site of the CMV-Cre vector (pBS185, Gibco/BRL) to generate pCMV-Cre-Puro (FIG. 8).
  • the pCMV-Cre- Puro vector was transfected by electroporation into A549E1-68 cells, and puromycin R (“puro R ”) clones were isolated. These puro R clones were then screened for expression of functional Cre recombinase.
  • the plasmid pBS/loxP-stop/MCLpA contains a lacZ cassette that is non-functional due to the presence of a stop codon (FIG. 9). This stop codon is surrounded by loxP sites, such that the propagation of this vector in a cell line producing Cre would excise the stop signal and activate the lacZ gene.
  • the pBS/loxP- stop/MCLpA vector was transiently transfected into each of the A549E1-Cre clones, and after 24 hours, the transfected cells were fixed and stained with X-Gal. LacZ expression of parental A549E1-68 cells (no Cre) was compared to lacZ expression in seven different puro r A549E1-Cre clones.
  • lacZ due to expression of Cre was observed as blue cells, at a frequency ranging from 1% to 50% in 20/26 puro R clones. This range of LacZ-expressing cells is most likely a reflection of the transient transfection efficiency of the different puro R clones with the pBS/loxPstop-MCLpA vector, although it could also reflect variations in Cre recombinase expression in different cell lines.
  • Western blot analysis using an anti-Cre antibody (Pharmingen) confirmed the presence of the 35 kd Cre protein in these cell lines.
  • KRAB Kruppel-associated box
  • the TetR-KRAB repressor binds to tetO sequences present in a transcriptional control region and represses transcription of genes placed as far as 3 kb downstream.
  • the present invention describes a system for tetracycline-controlled inhibition of helper virus packaging, comprising multiple tetO sequence in the helper virus packaging signal sequence, and an El helper cell line that constitutively expresses the TetR-KRAB protein.
  • the helper virus is still capable of replicating and providing all the necessary proteins, in trans, required for replication of the mini Ad vector, however, its packaging is attenuated due to binding of the TetR-KRAB protein to the tetO sites in the packaging signal.
  • the overall goal is to hinder or repress helper virus packaging, thus enriching for vector virus packaging.
  • This packaging repression is reversible, since in the presence of tetracycline, the TetR-KRAB repressor dissociates from the tetO sequences, and packaging is restored. Details of this tetO-controlled helper virus were presented in an earlier patent application (Serial No. 08/658,961, filed May 31, 1996).
  • the TetR-KRAB expressing cell line was derived using the A549E1-68 helper cell line described in Example 2.
  • A549E1-68 cells were transfected with a TetR-KRAB gene under control of the CMV promoter (see Deuschle et al., Mol. Cell. Biol. L5_ p. 1907-1914 (1995).
  • the TetR-KRAB vector also contains a hygromycin resistance gene for selection in mammalian cells.
  • a test vector see Deuschle, et al., Mol. and Cell. Biol. 15.
  • Hygromycin-resistant A549E1 -TetR-KRAB clones were transfected with pTetO-CMV-L by electroporation and each clone was split into two wells of a 6-well plate. 24 hours post-transfection, cells from one duplicate well were refed with medium containing tetracycline, and the other duplicate well in medium without tetracycline. After another 24 hours, cells were lysed and assayed for luciferase expression using a Promega Luciferase Assay Kit.
  • Two hygro R A549E1 clones (TKE-9 and TKE-12) demonstrated a 4 to 6 fold repression of luciferase reporter activity when grown in the absence of tetracycline versus cells grown in media containing Tet, indicating expression of the TetR-KRAB repressor protein in the cells (FIG. 10).
  • These A549E1 -TetR-KRAB cell lines will be used to test attenuation of the TetO-controlled Ad helper virus.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Virology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

A new series of helper cell lines for the complementation, amplification, and controlled attenuation of E1-deleted adenovirus are disclosed in the present invention. These cell lines are advantageous because they can complement adenovirus E1 genedeletions without production of replication competent adenovirus (RCA), thus making them safer for the large-scale production of adenovirus stock for use in human genetherapy trials. A preferred embodiment is an A549E1 cell line that contains only the Ad5 E1 DNA sequences sufficient for complementation of E1-deleted adenoviral vectors without sequences that overlap with the adenovirus vector. In another aspect, the present invention embodies methods for the production of second generation A549-E1 complementing cell lines that, in addition to producing E1, also produce proteins required for further manipulation of adenoviral vectors. A preferred embodiment is an A549E1 cell line with DNA sequences that encode a polypeptide sufficient for packaging attenuation of E1-deleted helper virus, in order to enrich for packaging of mini-adenovirus.

Description

ADENOVIRUS El -COMPLEMENTING CELL LINES
FIELD OF THE INVENTION
This invention relates to novel cells and methods for use in propagating El- deleted adeno viruses.
CROSS-REFERENCE This application is a continuation-in-part of serial number 08/658,961 filed May 31, 1996.
BACKGROUND OF THE INVENTION The majority of adenoviral vectors used in gene therapy applications were designed to have deletions in the El region of the adenovirus 5 (Ad5) genome. The El region, not including region IX, consists of 9% of the left end of Ad5 (1.2 - 9.8 map units), and is subdivided into two regions, E1A and E1B, each one coding for several proteins. Expression of E1A/E1B is required for virus replication and for expression of all other Ad5 proteins (E2-E4, Late Proteins; Ginsberg, H.S. The Adeno viruses. Plenum Press, New York. p.46-67(1984). Deletion of El, therefore creates a replication- incompetent virus that, in theory, is silent for expression of all Ad5 proteins and expresses only the transgene of interest. Deletion of El A and E1B is also of interest for safety reasons, since these two proteins, in combination, have been implicated in oncogenic transformation of mammalian cells (Graham, et al., In Cold Spring Harbor Symp. Quant. Biol. 39, p. 637-650 (1974); Van Der Eb, et al., Gene 2, p.l 15-132 (1977); McKinnon, et al., Gene 19, p. 33-42(1982). All of the Class I adenovirus vectors used to date in human clinical trials, are deleted for El.
El deficient adenoviral vectors are propagated in an Ad5 helper cell line called 293 (Graham, F.L. and Smiley, J, J. Gen. Virol. 36, p.54-72 (1977). 293 cells were derived by transforming human embryonic kidney cells with sheared fragments of Ad5 DNA. Genomic analysis revealed that 293 cells contain four to five copies per cell of the left 12% of the viral genome (including the entire El region) and approximately one copy per cell of 9% of the right end, the E4 region (Aiello,L.,et al, Virology 94, p.460-469 (1979). While 293 cells are very efficient at producing high titers of El -deficient adenovirus, they have the disadvantage that, due to the presence of Ad5 sequences besides El integrated into the 293 genome, recombination can occur with sequences in the El -deficient adenovirus vector causing the production of El -containing, replication- competent adenovirus (RCA). Depending on how early a passage the aberrant recombination event occurs during the amplification and propagation of the El -deficient adenovirus, and which passage is used for large-scale production of the adenovirus stock, production of RCA in 293 cells can present severe ramifications for the safety of human gene therapy trials (Lochmuller, H., et al., Human Gene Therapy 5., p. 1485-1491 (1994). In addition to production of RCA, recombination in 293 cells can also cause deletions and rearrangements that affect transgene expression, thereby decreasing the titer of functional adenovirus particles. Recently, cell lines have been developed using defined Ad5 DNA fragments, including the El region; however these cell lines contain significant sequence overlap with homologous sequences in the El -deleted adenovirus vectors, thus allowing for undesirable homologous recombination events and the possibility for generation of RCA (Fallaux, et al., Human Gene Therapy 1_, p. 215-222 (1996); Imler, et al, Gene Therapy 3, p. 75-84 (1996).
In this invention, a series of cell lines have been generated containing only the minimal El gene region for the complementation of El -deleted adenoviral vectors, in the absence of RCA.
SUMMARY OF THE INVENTION This invention encompasses a series of helper cell lines for the complementation, amplification, and controlled attenuation of El-deleted adenovirus. These cell lines are advantageous because they can complement adenovirus El gene deletions without production of replication competent adenovirus (RCA). A preferred embodiment is an A549E1 cell line that contains only the Ad5 El DNA sequences sufficient for complementation of El -deleted adenoviral vectors without sequences that overlap with the adenovirus vector. In a preferred embodiment, the El DNA sequences comprise El A and E1B genes.
In another aspect, the present invention embodies methods for selectively propagating mini-adenovirus without generating RCA, by transfecting an A549E1 cell line with DNA sequences that encode a polypeptide sufficient for packaging attenuation of El -deleted helper virus. In a preferred embodiment, the polypeptide comprises Cre recombinase. In another preferred embodiment, the polypeptide comprises TetR-KRAB.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 is a diagram indicating the structure of adenovirus sequences in a typical El- deleted Ad helper virus (top line), in 293 cells (ref.5), and in other El -containing cell lines, including 911 cells (ref.8) and A549E1-68 cells (this invention); and how recombination between homologous adenovirus sequences occurs to generate a replication-competent adenovirus (RCA).
Figure 2 is a diagram of the CMV-E1 mammalian expression vector.
Figure 3 is a Southern blot analysis of G418r A549E1 clones.
Figure 4A is a Western blot analysis of El A protein expression in A549 cells, 293 Cells, and A549El-68.
Figure 4B is the metabolic 35S labeling and immunoprecipitation of EIB proteins in
A549 Cells, 293 Cells and 4 single-cell clones derived from A549E1-68.
Figure 5 is a representation of the El -deleted adenovirus vector, Ad5-CA-GFP.
Figure 6 is an agarose gel analysis of 40 PCR reactions using ElA-specific primers for detection of RCA.
Figure 7 is a diagram of a system for the attenuation of helper virus with a loxP -modified packaging signal.
Figure 8 is a diagram of the pCMV-Cre-Puro vector.
Figure 9 is a diagram of the pBS/loxP-stop/MCLpA vector. Figure 10 is a bar graph depicting luciferase expression in both control cells (A549E1-
68) and cell lines expressing the TetR-KRAB protein, following transient transfection with the pTetO7-CMN-L test vector.
DETAILED DESCRIPTION OF THE INVENTION This invention provides cell lines that can complement El -deleted adenovirus without the disadvantage of undesirable recombination and RCA. These cell lines are obtained by cloning and expressing in the A549 cell line only those sequences that are required for El complementation and excluding from the cell line all other Ad5 sequences that have homology to the vector and could cause recombination to produce RCA.
Currently 293 cells are used to propagate El -deleted adenovirus. However, 293 cells harbor DNA fragments that make up 21 % of the Ad5 genome. 293 cells therefore have significant sequence homology (outside of the El region) that overlaps with sequences in the adenoviral vectors, which can allow for homologous recombination events to produce a wild type adenoviral particle. FIG. 1 shows the structure of adenovirus sequences in 293 cells versus other El -containing cell lines (including A549E1 cells), and how recombination between homologous adenovirus sequences occurs to generate a replication-competent adenovirus (RCA). Significant homology (more than 1000 base pairs) exists between Ad sequences 3' to the El region in conventional adenovirus vectors, and sequences integrated into the helper cell genome of 293 cells and 911 cells. Reciprocal recombination across these homologous regions can result in the generation of a wild-type, El(+), replication-competent adenovirus (RCA). This invention embodies the cloning of an El A and ElB-encoding DNA fragment from Ad5 into a mammalian expression vector. This El vector was stably transfected into human A549 cells to produce an El expression cell line. The genome of a representative cell line, A549E1-68, contains no sequence overlap with sequences present in the El -deleted Ad helper virus and thus, recombination to produce RCA is not possible (FIG. 1). Characterization of this A549E1 cell line demonstrated the production of El A and EIB proteins, high infectivity with adenovirus vectors, complementation of El- deleted adenovirus to produce high-titer virus stocks, as well as, the lack of production of replication-competent adenovirus (RCA). Further embodiments of the invention are described for the production of 2nd generation A549-E1 complementing cell lines which, in addition to producing El, also produce proteins required for further manipulation of adenoviral vectors, thus providing a novel series of RCA-free adenovirus helper cell lines, as tools for novel virus production. The first example is an A549E1 cell line which expresses the Cre recombinase. This invention provides a novel El -deleted helper virus whose packaging signal is flanked by loxP sites, and when this helper virus is propagated in an A549E1 cell line expressing Cre, the packaging signal is deleted by excision, thus attenuating helper virus packaging and enriching for packaging of mini-adenovirus (Ad5 virus which is devoid of all viral protein-coding sequences). This is advantageous because during the production of helper-dependent, mini-adenovirus it becomes necessary to attenuate helper virus packaging in order to enrich for the mini-virus. 293-Cre cells have been generated for this purpose (Parks, R. , et al, P.N.A.S. 93, p.13565-13570 (1996), however, A549E1- Cre cells have an advantage in that they would perform this task in an RCA-free environment.
A further embodiment of the present invention includes an A549-E1 complementing cell line which expresses the TetR-KRAB fusion protein, which would be used to amplify, and control the packaging efficiency of an El -deleted helper virus whose packaging signal has been modified to contain multiple tetracycline operator (tetO) sites. When this helper virus is propagated in an A549E1 cell line expressing TetR-KRAB, the repressor binds to the tetO sequence and specifically attenuates helper virus packaging, thus enriching for packaging of the mini-adenovirus which has a normal, wild-type packaging signal. Packaging of the helper virus can be restored by growing the cells and virus in the presence of tetracycline, which binds to the tet-KRAB repressor causing its dissociation from the tetO/packaging signal and a reversal of packaging repression. Detailed examples involving the derivation, characterization, and applications of these El -complementing helper cell lines are described in the following sections.
Example 1 Construction of the E1A/E1B vector To generate an expression vector which harbors only the E1A/E1B sequences required for complementation, a 3.1 kb DNA fragment coding for Ad5 El A and EIB genes was cloned in two pieces, sequentially, into the superlinker vector, pSL301 (Invitrogen). First, an 881 bp Afl III to Xbal fragment (Ad5 base pairs 462-1343) was cloned from pBRXad5KpnICl (a subclone of pJM17) into pSL301 (Afl III/XbaI). Second, a contiguous 2194 bp Xbal to Afl II (Ad5 base pairs 1343-3537) was cloned from pBRXad5XhoIClinto the same vector. The resultant 3075 bp El fragment (in pSL301) contains the TATA box and RNA cap site for El A, El A coding sequence, complete EIB promoter, and EIB coding sequence, including the stop codon for EIB p55 protein, but not including region IX. The 3075 bp Afl III - Afl II E1A/E1B fragment (Ad5 base pairs 462-3537) was isolated, blunt-ended with Klenow enzyme, and blunt-end ligated into the EcoRV site of the mammalian expression vector, pCDNA3 (Invitrogen), under control of the CMV promoter/enhancer. This process generated an Ad5-El expression vector, pCMV-El (FIG.2). Example 2 Generation and characterization of the El cell line
This CMV-El expression plasmid, (FIG. 2), was transfected using Lipofectamine (Gibco BRL) into A549 human lung carcinoma cells (ATCC CRL 185) and G418R colonies were isolated. Single-cell clones were screened for functional E1A/E1B expression. An El -deleted adenovirus containing a green florescence protein (GFP) expression cassette, Ad5 CA-GFP, was used to infect the A549-E1 clones. Three days post-infection, clones were screened for production of El -complemented Ad5 CA-GFP adenovirus by visual examination for cytopathic effect (CPE). One clone, A549E1-68, displayed 100% CPE in 3 days, similar to that observed for 293 cells. The clear area in the center of the plaque is evidence of CPE caused by El -complemented virus amplification. This clone also showed high infectivity, in that virtually 100% of the cells fluoresced green 24 hours post-infection. The high infection rate and rapid generation of CPE induced in this cell line is strong evidence that functional E1A/E1B proteins are being produced that are capable of promoting replication and amplification of the El- deleted Ad5-CA-GFP virus. The A459E1 cell was deposited at the American Type Tissue Culture Collection (ATCC) under the Budapest Treaty on January 15, 1998 as ATCC Designation CRL-12458 (viability confirmed January 20, 1998).
FIG. 3 shows a Southern blot using an El sequence-specific DNA probe. This assay demonstrated the presence of the CMV-El transgene in A549E1-68 (Lane 4), and a subclone of A549E1-68 (El-68.3), but not in the parental A549 cell line (Lane 2). Sequences hybridizing with the El -specific probe were also observed in 293 cells as expected since they complement El-deleted adenovirus (Lane 3). The morphology of the El-transfected cells was significantly different from the parental A549 cell line. A549 cells at sub-confluent density grow as distinct single cells with an elongated, fibroblast- like morphology, whereas the El cell line A549E1-68 grows as colonies of cells with a more cuboidal morphology. A549E1-68 was also compared with 293 cells for production of El -deleted adenovirus (Ad5 CA-GFP) by plaque assay and found to produce an equivalent titer of complemented virus (7 x 109 pfu for A549E1-68 vs. 9 x 109 pfu for 293).
FIG. 4A shows a Western blot analysis using an EIA specific antibody (M73, Oncogene Science). This antibody detected two ElA-specific bands with apparent molecular weights of 46kd and 42kd in the A549E1-68 cell line (lane 3), corresponding to products expected from EIA 13S and 12S mRNAs (Ginsberg, 1984), and identical in size to those observed in 293 cells (lane 2). These El A-specific bands were not detected in parental A549 cells (lane 1). FIG. 4B shows the immunoprecipitation of metabolically-radiolabeled proteins by a monoclonal antibody specific for EIB p55. A549E1-68 produced an immunoreactive band of approximately 55 kd (lane 3) that was not detected in parental A549 cells. This 55 kd, ElB-specific band, as well as secondary background bands, were observed in 293 cells also (lane 2). Extra "background" bands found in both experimental and control lanes have been observed by other authors and are attributed to co-immunoprecipitation of a variety of proteins including, cyclins, p53, and Rb. It is clear that A549E1-68 not only expresses EIA and EIB, but that they are functional, since this cell line can complement for production of high titer, El -deleted, recombinant adenovirus. Example 3 El-deleted adenovirus produced in A549E1 cells is RCA-free
To prove that this new Ad5 helper cell line can complement without production of RCA, a series of PCR RCA assays were performed following amplification in A549E1 cells of the E 1 -deleted Ad5 -CA-GFP adenovirus vector. The Ad5-CA-GFP vector is illustrated in FIG. 5. It contains a transcriptional control element consisting of the CMV
enhancer and the β-actin promoter and the deletion of El sequences includes a lack of
Ad5 DNA through base pair 3550. Since the E1A/E1B complementing region contained in the A549E1 cells extends only to base pair 3537, there is no overlapping sequence homology to allow RCA production. El -deleted vectors with smaller deletions may still contain some El DNA and should be avoided, as they could still allow RCA to occur at a low frequency.
For the PCR RCA assay, Ad5-CA-GFP virus was serially propagated through 20 passages on A549E1-68 cells. Following serial propagation and virus amplification, Ad5-CA-GFP virus DNA was isolated by freeze-thaw lysis, and PCR was performed using primers specific for either the EIA region or the E2B region. Amplification of an 880 bp E2B product serves as a PCR positive control, while the presence of a 1086 bp El A-specific product is evidence that an El (+) replication-competent adenovirus (RCA) has been produced during amplification of the El (-) Ad5 -CA-GFP. 20 ug of Ad5-CA- GFP virus DNA (equivalent to 1 x 1010 virus particles), obtained from amplification in A549E1 cells, was divided into 40 PCR reactions and tested for RCA using the EIA primers (FIG. 6). For both top and bottom panels of FIG. 6, lane 1 contains 1 kb DNA markers, lane 2 contains wild type Ad5 virus DNA, lane 3 consists of PCR of Ad5-CA- GFP virus DNA (E1-) isolated from 20+ passages on A549E1-68 cells using EIA and E2B specific primers (positive control), and lanes 4-20 consist of PCR of Ad5-CA-GFP virus DNA (E1-) isolated from 20+ passages on A549E1-68 cells, using ElA-specific primers only. No 1086 bp El region specific PCR fragments were detected in any of the reactions indicating that no RCA was present in the virus prep.
A second, CPE-based RCA assay was performed by amplifying El -deleted adenovirus (Ad5-CA-GFP) on A549E1-68 cells and testing the amplified virus by passaging, on normal A549 cells (don't make El) for production of El -containing RCA. Plaque formation (CPE) on a monolayer of normal A549 cells would provide evidence for the production of wild-type (El +) virus during amplification on the El helper cell line, A549E1-68. 2 x 1010 El (-) virus particles (amplified using A549E1-68) were used to infect each of five 150mm plates of normal A549 cells (1 x 10" particles total). No CPE or single plaques were detected after 8 days on any of the A549 plates, indicating the absence of any El (+) RCA virus in the A549El-68-amplified virus prep. Therefore, using two sensitive assays for detection of RCA, no wild-type recombinant El (+) virus was detected, supporting the utility of this cell line for the amplification and large-scale preparation of El -deleted adenoviral vectors in the absence of RCA.
Example 4 Generation of an A549E1 Cre Cell Line
A 2nd generation El -complementing cell line was generated using the A549E1- 68.3 clonal line for transfection with Cre recombinase. This cell line will both complement El -deleted adenovirus vectors and mediate the excision of sequences surrounded by loxP sites. Our primary use for this cell line is to further attenuate packaging of an Ad5 helper virus, whose packaging signal is flanked by two loxP sites (FIG. 7), in order to enrich for packaging of the desired El -deficient, mini-adenovirus vector. 293 cells expressing the Cre recombinase were generated for a similar purpose by Parks et al. (P.N.A.S. 93:13565-13570), and were shown to increase the titer of El- deleted vector virus 10 fold per passage, demonstrating the overall utility of this system for removal of helper virus. The A549E1-Cre cell line described in this invention will not only attenuate helper virus packaging in a similar fashion, it also has the advantage that any adenovirus produced will be free of deleterious RCA. As a first step towards the production of the A549E1-Cre cell line, a Cre expression vector was constructed. A 1440 bp SV40 promo ter-puromycin cassette (for selection in NeoR A549E1 cells) was cloned into a unique EcoRI site of the CMV-Cre vector (pBS185, Gibco/BRL) to generate pCMV-Cre-Puro (FIG. 8). The pCMV-Cre- Puro vector was transfected by electroporation into A549E1-68 cells, and puromycinR ("puroR") clones were isolated. These puroR clones were then screened for expression of functional Cre recombinase. The plasmid pBS/loxP-stop/MCLpA contains a lacZ cassette that is non-functional due to the presence of a stop codon (FIG. 9). This stop codon is surrounded by loxP sites, such that the propagation of this vector in a cell line producing Cre would excise the stop signal and activate the lacZ gene. The pBS/loxP- stop/MCLpA vector was transiently transfected into each of the A549E1-Cre clones, and after 24 hours, the transfected cells were fixed and stained with X-Gal. LacZ expression of parental A549E1-68 cells (no Cre) was compared to lacZ expression in seven different puror A549E1-Cre clones. Expression of lacZ (due to expression of Cre) was observed as blue cells, at a frequency ranging from 1% to 50% in 20/26 puroR clones. This range of LacZ-expressing cells is most likely a reflection of the transient transfection efficiency of the different puroR clones with the pBS/loxPstop-MCLpA vector, although it could also reflect variations in Cre recombinase expression in different cell lines. Western blot analysis using an anti-Cre antibody (Pharmingen), confirmed the presence of the 35 kd Cre protein in these cell lines. Experiments to assess the attenuation of Ad helper virus containing a packaging signal flanked by two loxP sites are in progress.
Example 5 Generation of an A549E1 cell line expressing TetR-KRAB
A highly conserved, 75 amino acid protein called the Kruppel-associated box (KRAB) was recently isolated by Margolin, et al. (P.N.A.S. 91, p.4509-4513 (1994)). The KRAB box is a member of the Kox-1 family of human zinc finger proteins and was subsequently shown to be a strong transcriptional repressor. By fusing the KRAB domain from Kox-1 to the Tet repressor derived from TnlO oϊ Escherichia coli, a hybrid protein was generated, TetR-KRAB, which allows tetracycline-controlled silencing of eukaryotic promoters (Deuschle, et al., Mol. and Cell. Biol. 1J p.1907-1914 (1995). The TetR-KRAB repressor binds to tetO sequences present in a transcriptional control region and represses transcription of genes placed as far as 3 kb downstream. The present invention describes a system for tetracycline-controlled inhibition of helper virus packaging, comprising multiple tetO sequence in the helper virus packaging signal sequence, and an El helper cell line that constitutively expresses the TetR-KRAB protein. The helper virus is still capable of replicating and providing all the necessary proteins, in trans, required for replication of the mini Ad vector, however, its packaging is attenuated due to binding of the TetR-KRAB protein to the tetO sites in the packaging signal. The overall goal is to hinder or repress helper virus packaging, thus enriching for vector virus packaging. This packaging repression is reversible, since in the presence of tetracycline, the TetR-KRAB repressor dissociates from the tetO sequences, and packaging is restored. Details of this tetO-controlled helper virus were presented in an earlier patent application (Serial No. 08/658,961, filed May 31, 1996).
The TetR-KRAB expressing cell line was derived using the A549E1-68 helper cell line described in Example 2. A549E1-68 cells were transfected with a TetR-KRAB gene under control of the CMV promoter (see Deuschle et al., Mol. Cell. Biol. L5_ p. 1907-1914 (1995). The TetR-KRAB vector also contains a hygromycin resistance gene for selection in mammalian cells. A test vector (see Deuschle, et al., Mol. and Cell. Biol. 15. p.1907- 1914 (1995)) that has a luciferase reporter gene under control of the CMV promoter fused to a TetO sequence was used for transient transfection into cells lacking a TetR-KRAB repressor. This transfection results in high level expression of luciferase, whereas transfection into A549E1 cells expressing the TetR-KRAB protein will result in the repression of luciferase due to binding of the repressor to tetO sites in the test vector. Hygromycin-resistant A549E1 -TetR-KRAB clones were transfected with pTetO-CMV-L by electroporation and each clone was split into two wells of a 6-well plate. 24 hours post-transfection, cells from one duplicate well were refed with medium containing tetracycline, and the other duplicate well in medium without tetracycline. After another 24 hours, cells were lysed and assayed for luciferase expression using a Promega Luciferase Assay Kit. Two hygroR A549E1 clones (TKE-9 and TKE-12) demonstrated a 4 to 6 fold repression of luciferase reporter activity when grown in the absence of tetracycline versus cells grown in media containing Tet, indicating expression of the TetR-KRAB repressor protein in the cells (FIG. 10). These A549E1 -TetR-KRAB cell lines will be used to test attenuation of the TetO-controlled Ad helper virus. These examples are intended to illustrate the present invention and are not intended to limit it in spirit or scope.

Claims

CLAIMSWE CLAIM:
1. A recombinant cell, comprising a mammalian cell capable of expressing a region of the adenoviral El sequence sufficient for complementation of El -deleted adenoviral vectors without generating replication-competent adenovirus.
2. The recombinant cell of claim 1 further comprising said mammalian cell capable of expressing DNA sequences that encode a polypeptide sufficient for attenuation control of El -deleted helper virus.
3. The recombinant cell of claim 2 wherein said DNA sequences comprise a Cre recombinase-encoding DNA.
4. The recombinant cell of claim 2 wherein said DNA sequences comprise a TetR- KRAB-encoding DNA.
5. A method for propagating El -deleted adenovirus without generating replication- competent adenovirus comprising: (a) transfecting a mammalian cell with a region of the adenoviral El sequence sufficient for complementation of said El -deleted adenovirus without generating replication-competent adenovirus;
(b) infecting said mammalian cells with said El -deleted adenovirus; and
(c) growing said mammalian cells under conditions suitable for lysis of said mammalian cells by said complementation of El -deleted adenovirus.
6. A method of selectively propagating mini-adenovirus without generating replication-competent adenovirus comprising:
(a) transfecting a mammalian cell with a region of the adenoviral El sequence sufficient for complementation of said El -deleted adenovirus without generating replication-competent adenovirus; (b) further transfecting said mammalian cell with DNA sequences that encode a polypeptide sufficient for attenuation control of El -deleted helper virus; (c) co-infecting said mammalian cells with said El -deleted helper virus, said El- deleted helper virus containing a modified packaging signal that is controlled by said polypeptide; (d) growing said mammalian cells under conditions sufficient for lysis of said mammalian cells by said complementation of El -deleted adenovirus; and for said attenuation control of said E 1 -deleted helper virus.
7. The method of claim 6, wherein
(a) said DNA sequences that encode a polypeptide sufficient for attenuation control of El -deleted helper virus comprise a Cre recombinase-encoding DNA; and (b) said modified packaging signal that is controlled by said polypeptide sufficient for attenuation of El -deleted helper virus comprises loxP sites adjacent to said packaging signal.
8. The method of claim 6, wherein
(a) said DNA sequences that encode a polypeptide sufficient for attenuation control of El -deleted helper virus comprise a TetR-KRAB-encoding DNA; and
(b) said modified packaging signal that is controlled by said protein for attenuation of El -deleted helper virus comprises a tetO DNA sequence in said modified packaging signal.
9. A method for making a recombinant cell comprising:
(a) transfecting a mammalian cell with a region of the adenoviral El sequence sufficient for complementation of El -deleted adenoviral vectors without generating replication-competent adenovirus; and
(b) selecting for recombinant cells that express said region of the adenoviral El gene.
10. A method for using a recombinant mammalian cell capable of expressing a region of the adenoviral El gene sufficient for complementation of El -deleted adenovirus without generating replication-competent adenovirus, comprising:
(a) infecting said recombinant mammalian cells with said El -deleted adenovirus; and
(b) growing said mammalian cells under conditions sufficient for lysis of said mammalian cells by said complementation of El -deleted adenovirus and propagation of said El -deleted adenovirus.
11. The method of claim 10 wherein said recombinant mammalian cell is a human cell.
12. The method of claim 10 wherein said recombinant mammalian cell is A549 human lung carcinoma cell.
13. The method of claim 10 wherein said recombinant mammalian cell is deposited with the ATCC under accession number CRL-12458.
14. A method of using a recombinant cell, comprising a mammalian cell capable of expressing adenoviral EIA and EIB sequences sufficient for complementation of El -deleted adenovirus without generating replication-competent adenovirus, comprising:
(a) infecting said recombinant mammalian cells with said El -deleted adenovirus; and (b) growing said mammalian cells under conditions sufficient for lysis of said mammalian cells by said complementation of El -deleted adenovirus and propagation of said El -deleted adenovirus.
15. The method of claim 14 wherein said recombinant mammalian cell is a human cell.
16. The method of claim 14 wherein said recombinant mammalian cell is A549 human lung carcinoma cell.
17. The method of claim 14 wherein said recombinant mammalian cell is deposited with the ATCC under accession number CRL-12458.
18. A method for using a recombinant mammalian cell capable of expressing a region of the adenoviral El gene sufficient for complementation of El -deleted helper virus without generating replication-competent adenovirus and also capable of expressing DNA sequences that encode a protein for attenuation control of said
El -deleted helper virus, comprising:
(a) co-infecting said mammalian cells with an El -deleted helper virus, said El- deleted helper virus containing a modified packaging signal that is controlled by said protein for attenuation of El -deleted adenovirus, and a mini-
adenovirus; and (b) growing said mammalian cells under conditions sufficient for lysis of said mammalian cells by said complementation of El -deleted adenovirus and also sufficient for said attenuation control of said El -deleted helper virus.
19. The method of claim 18 wherein said recombinant mammalian cell is a human cell.
20. The method of claim 18 wherein said recombinant mammalian cell is A549 human lung carcinoma cell.
21. The method of claim 18 wherein said DNA sequences comprise a Cre recombinase-encoding DNA that encodes a polypeptide sufficient for said attenuation control of E 1 -deleted adenovirus.
22. The method of claim 18 wherein said recombinant mammalian cell is a human cell.
23. The method of claim 18 wherein said recombinant mammalian cell is A549 human lung carcinoma cell.
24. The method of claim 18 wherein said recombinant mammalian cell is deposited with the ATCC under accession number CRL-12458.
25. The method of claim 18 wherein said DNA sequences comprise a TetR-KRAB- encoding DNA that encodes a polypeptide sufficient for said attenuation control of El -deleted adenovirus.
26. The method of claim 25 wherein said recombinant mammalian cell is a human cell.
27. The method of claim 25 wherein said recombinant mammalian cell is A549 human lung carcinoma cell.
28. The method of claim 25 wherein said recombinant mammalian cell is deposited with the ATCC under accession number CRL-12458.
29. A method for using a recombinant mammalian cell capable of expressing adenoviral EIA and EIB sequences sufficient for complementation of El -deleted helper virus without generating replication-competent adenovirus and also capable of expressing a protein for controlling attenuation of said El -deleted helper virus, comprising :
(a) co-infecting said mammalian cells with an El -deleted helper virus, said El -deleted helper virus containing a modified packaging signal that is controlled by said protein for attenuation of El -deleted adenovirus, and a mini-adenovirus; and
(b) growing said mammalian cells under conditions sufficient for lysis of said mammalian cells by said complementation of El -deleted adenovirus and also sufficient for said attenuation control of said El -deleted helper virus.
30. The method of claim 29 wherein said recombinant mammalian cell is a human cell.
31. The method of claim 29 wherein said recombinant mammalian cell is A549 human lung carcinoma cell.
32. The method of claim 29 wherein said DNA sequences comprise a Cre recombinase-encoding DNA that encodes a polypeptide sufficient for said attenuation control of El-deleted adenovirus.
33. The method of claim 32 wherein said recombinant mammalian cell is a human cell.
34. The method of claim 32 wherein said recombinant mammalian cell is A549 human lung carcinoma cell.
35. A method of claim 32 wherein said recombinant mammalian cell is deposited with the ATCC under accession number CRL-12458.
36. A method of claim 29 wherein said DNA sequences comprise a TetR-KRAB- encoding DNA that encodes a polypeptide sufficient for said attenuation control of El -deleted adenovirus.
37. A method of claim 36 wherein said recombinant mammalian cell is a human cell.
38. A method of claim 36 wherein said recombinant mammalian cell is A549 human lung carcinoma cell.
39. The method of claim 38 wherein said recombinant mammalian cell is deposited with the ATCC under accession number CRL-12458.
EP98908661A 1997-03-04 1998-02-23 Adenovirus e1-complementing cell lines Withdrawn EP0973866A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US81003997A 1997-03-04 1997-03-04
US810039 1997-03-04
PCT/US1998/003473 WO1998039411A1 (en) 1997-03-04 1998-02-23 Adenovirus e1-complementing cell lines

Publications (2)

Publication Number Publication Date
EP0973866A1 true EP0973866A1 (en) 2000-01-26
EP0973866A4 EP0973866A4 (en) 2000-04-19

Family

ID=25202818

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98908661A Withdrawn EP0973866A4 (en) 1997-03-04 1998-02-23 Adenovirus e1-complementing cell lines

Country Status (4)

Country Link
EP (1) EP0973866A4 (en)
JP (1) JP2000509614A (en)
CA (1) CA2283253A1 (en)
WO (1) WO1998039411A1 (en)

Families Citing this family (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7604960B2 (en) 1999-04-15 2009-10-20 Crucell Holland B.V. Transient protein expression methods
US7297680B2 (en) 1999-04-15 2007-11-20 Crucell Holland B.V. Compositions of erythropoietin isoforms comprising Lewis-X structures and high sialic acid content
WO2003048348A2 (en) 2001-12-07 2003-06-12 Crucell Holland B.V. Production of viruses, viral isolates and vaccines
US6855544B1 (en) 1999-04-15 2005-02-15 Crucell Holland B.V. Recombinant protein production in a human cell
US8236561B2 (en) 1999-04-15 2012-08-07 Crucell Holland B.V. Efficient production of IgA in recombinant mammalian cells
US7527961B2 (en) 1999-11-26 2009-05-05 Crucell Holland B.V. Production of vaccines
US7521220B2 (en) 1999-11-26 2009-04-21 Crucell Holland B.V. Production of vaccines
US7192759B1 (en) 1999-11-26 2007-03-20 Crucell Holland B.V. Production of vaccines
AU2003290453A1 (en) 2002-12-20 2004-07-14 Chromagenics B.V. Means and methods for producing a protein through chromatin openers that are capable of rendering chromatin more accessible to transcription factors
WO2004099396A1 (en) 2003-05-09 2004-11-18 Crucell Holland B.V. Cultures of e1-immortalized cells and processes for culturing the same to increase product yields therefrom
AU2005214090B2 (en) 2004-02-23 2008-09-11 Crucell Holland B.V. Virus purification methods
WO2005123918A1 (en) * 2004-06-22 2005-12-29 Tokai University Educational System Expression vector capable of regulating the induction of foreign gene expression
US20060195935A1 (en) 2004-11-08 2006-08-31 Chromagenics B.V. Selection of host cells expressing protein at high levels
US8999667B2 (en) 2004-11-08 2015-04-07 Chromagenics B.V. Selection of host cells expressing protein at high levels
ES2384391T3 (en) 2004-11-08 2012-07-04 Chromagenics B.V. Selection of host cells that express protein at high levels
US8039230B2 (en) 2004-11-08 2011-10-18 Chromagenics B.V. Selection of host cells expressing protein at high levels
BRPI0517380A (en) 2004-11-08 2008-10-07 Chromagenics Bv DNA molecule, expression cassette, host cell, and methods for generating a host cell expressing a polypeptide of interest and for producing a polypeptide of interest
CN101155915B (en) 2005-04-11 2013-12-04 克鲁塞尔荷兰公司 Virus purification using ultrafiltration
ES2532015T3 (en) 2008-11-03 2015-03-23 Crucell Holland B.V. Method for the production of adenoviral vectors
AU2010305765B2 (en) 2009-10-15 2015-07-02 Crucell Holland B.V. Method for the purification of adenovirus particles
CN102575233B (en) 2009-10-15 2014-07-16 克鲁塞尔荷兰公司 Process for adenovirus purification from high cell density cultures
WO2011098592A1 (en) 2010-02-15 2011-08-18 Crucell Holland B.V. Method for the production of ad26 adenoviral vectors
US8932607B2 (en) 2012-03-12 2015-01-13 Crucell Holland B.V. Batches of recombinant adenovirus with altered terminal ends
CN104379733B (en) 2012-03-12 2016-01-20 克鲁塞尔荷兰公司 Tool changes the recombinant adenovirus group of end
MY169352A (en) 2012-03-22 2019-03-25 Janssen Vaccines & Prevention Bv Vaccine against rsv
US9125870B2 (en) 2012-03-22 2015-09-08 Crucell Holland B.V. Vaccine against RSV
MX361774B (en) 2013-04-25 2018-12-17 Janssen Vaccines & Prevention Bv Stabilized soluble prefusion rsv f polypeptides.
PE20160045A1 (en) 2013-06-17 2016-02-18 Crucell Holland Bv SOLUBLE AND STABILIZED RESPIRATORY SYNCITIAL VIRUS (RSV) PREFUSION POLYPEPTIDES F
CN107075521B (en) 2014-11-04 2021-06-08 扬森疫苗与预防公司 Therapeutic HPV16 vaccine
US10570417B2 (en) 2015-04-14 2020-02-25 Janssen Vaccines & Prevention B.V. Recombinant adenovirus expressing two transgenes with a bidirectional promoter
JP6840718B2 (en) 2015-07-07 2021-03-10 ヤンセン ファッシンズ アンド プリベンション ベーフェーJanssen Vaccines & Prevention B.V. Stabilized pre-soluble fusion RSV F polypeptide
WO2017005844A1 (en) 2015-07-07 2017-01-12 Janssen Vaccines & Prevention B.V. Vaccine against rsv
EA037295B1 (en) 2015-08-20 2021-03-05 Янссен Вэксинс Энд Превеншн Б.В. Therapeutic hpv18 vaccines
AU2017248021B2 (en) 2016-04-05 2021-08-12 Janssen Vaccines & Prevention B.V. Stabilized soluble pre-fusion RSV F proteins
BR112018070323A2 (en) 2016-04-05 2019-01-29 Janssen Vaccines & Prevention Bv rsv vaccine
AU2017259259B2 (en) 2016-05-02 2020-11-19 Bavarian Nordic A/S Therapeutic HPV vaccine combinations
WO2017207480A1 (en) 2016-05-30 2017-12-07 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion rsv f proteins
US11001858B2 (en) 2016-06-20 2021-05-11 Janssen Vaccines & Prevention B.V. Potent and balanced bidirectional promoter
WO2018011196A1 (en) 2016-07-14 2018-01-18 Janssen Vaccines & Prevention B.V. Hpv vaccines
KR102111244B1 (en) 2017-02-09 2020-05-15 얀센 백신스 앤드 프리벤션 비.브이. Powerful short promoter for expression of heterologous genes
EP3613849A4 (en) * 2017-04-21 2021-01-06 Geneuin-Tech Co., Ltd. Cell line for producing nonreplicating adenovirus, and preparation method therefor
EP3624844A1 (en) 2017-05-17 2020-03-25 Janssen Vaccines & Prevention B.V. Methods and compositions for inducing protective immunity against rsv infection
EP3681533A1 (en) 2017-09-15 2020-07-22 Janssen Vaccines & Prevention B.V. Method for the safe induction of immunity against rsv
US11142551B2 (en) 2017-10-31 2021-10-12 Janssen Vaccines & Prevention B.V. Adenovirus and uses thereof
SG11202003398SA (en) 2017-10-31 2020-05-28 Janssen Vaccines & Prevention Bv Adenovirus vectors and uses thereof
SG11202003290RA (en) 2017-10-31 2020-05-28 Janssen Vaccines & Prevention Bv Adenovirus and uses thereof
WO2019086450A1 (en) 2017-10-31 2019-05-09 Janssen Vaccines & Prevention B.V. Adenovirus and uses thereof
TW202043256A (en) 2019-01-10 2020-12-01 美商健生生物科技公司 Prostate neoantigens and their uses
CA3140234A1 (en) 2019-05-15 2020-11-19 Janssen Vaccines & Prevention B.V. Prophylactic treatment of respiratory syncytial virus infection with an adenovirus based vaccine
US20220273787A1 (en) 2019-05-15 2022-09-01 Janssen Vaccines & Prevention B.V. Co-administration of seasonal influenza vaccine and an adenovirus based respiratory syncytial virus vaccine
IL291852A (en) 2019-10-03 2022-06-01 Janssen Vaccines & Prevention Bv Adenovirus vectors and uses thereof
AU2020385683A1 (en) 2019-11-18 2022-06-30 Janssen Biotech, Inc. Vaccines based on mutant CALR and JAK2 and their uses
TW202144388A (en) 2020-02-14 2021-12-01 美商健生生物科技公司 Neoantigens expressed in ovarian cancer and their uses
TW202144389A (en) 2020-02-14 2021-12-01 美商健生生物科技公司 Neoantigens expressed in multiple myeloma and their uses
JP2023521194A (en) 2020-04-13 2023-05-23 ヤンセン バイオテツク,インコーポレーテツド PSMA and STEAP1 vaccines and their uses
US20230024133A1 (en) 2020-07-06 2023-01-26 Janssen Biotech, Inc. Prostate Neoantigens And Their Uses
EP4175664A2 (en) 2020-07-06 2023-05-10 Janssen Biotech, Inc. Prostate neoantigens and their uses
US20230035403A1 (en) 2020-07-06 2023-02-02 Janssen Biotech, Inc. Method For Determining Responsiveness To Prostate Cancer Treatment

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997000326A1 (en) 1995-06-15 1997-01-03 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5919676A (en) * 1993-06-24 1999-07-06 Advec, Inc. Adenoviral vector system comprising Cre-loxP recombination
DE69535178T2 (en) * 1994-06-10 2006-12-14 Genvec, Inc. ADENOVER VECTOR SYSTEMS AND CELL LINES
IL116816A (en) * 1995-01-20 2003-05-29 Rhone Poulenc Rorer Sa Cell for the production of a defective recombinant adenovirus or an adeno-associated virus and the various uses thereof
CA2177085C (en) * 1996-04-26 2007-08-14 National Research Council Of Canada Adenovirus e1-complementing cell lines

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997000326A1 (en) 1995-06-15 1997-01-03 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of WO9839411A1

Also Published As

Publication number Publication date
EP0973866A4 (en) 2000-04-19
JP2000509614A (en) 2000-08-02
WO1998039411A1 (en) 1998-09-11
CA2283253A1 (en) 1998-09-11

Similar Documents

Publication Publication Date Title
EP0973866A1 (en) Adenovirus e1-complementing cell lines
WO1998039411A9 (en) Adenovirus e1-complementing cell lines
Zhang et al. Role for the adenovirus IVa2 protein in packaging of viral DNA
Massie et al. Inducible overexpression of a toxic protein by an adenovirus vector with a tetracycline-regulatable expression cassette
US6228646B1 (en) Helper-free, totally defective adenovirus for gene therapy
Holterman et al. Novel replication-incompetent vector derived from adenovirus type 11 (Ad11) for vaccination and gene therapy: low seroprevalence and non-cross-reactivity with Ad5
JP3492700B2 (en) Adenovirus vectors for gene therapy
US6365394B1 (en) Cell lines and constructs useful in production of E1-deleted adenoviruses in absence of replication competent adenovirus
US6140087A (en) Adenovirus vectors for gene therapy
IL160406A (en) Cell harbouring nucleic acid encoding adenovirus e1a and e1b gene products
US20020037280A1 (en) Recombinant, modified adenoviral vectors for tumor specific gene expression and uses thereof
Murakami et al. An adenoviral vector expressing human adenovirus 5 and 3 fiber proteins for targeting heterogeneous cell populations
Catalucci et al. An adenovirus type 5 (Ad5) amplicon-based packaging cell line for production of high-capacity helper-independent ΔE1-E2-E3-E4 Ad5 vectors
US20010046965A1 (en) Adenovirus E1-complementing cell lines
Wang et al. Episomal segregation of the adenovirus enhancer sequence by conditional genome rearrangement abrogates late viral gene expression
CA2318737A1 (en) Methods for pseudoadenoviral vector production
Bernt et al. A new type of adenovirus vector that utilizes homologous recombination to achieve tumor-specific replication
Gall et al. Rescue and production of vaccine and therapeutic adenovirus vectors expressing inhibitory transgenes
US20060210965A1 (en) Efficient generation of adenovirus-based libraries by positive selection of adenoviral recombinants through ectopic expression of the adenovirus protease
Fang et al. Diminishing adenovirus gene expression and viral replication by promoter replacement
EP1127149B1 (en) Cells for the production of helper dependent adenoviral vectors
EP1291434A2 (en) Efficient generation of adenovirus-based libraries by positive selection of adenoviral recombinants through ectopic expression of the adenovirus protease
McVoy et al. Tetracycline-mediated regulation of gene expression within the human cytomegalovirus genome
JP2004517610A (en) Self-rearranging DNA vector
Howe et al. Matching complementing functions of transformed cells with stable expression of selected viral genes for production of E1-deleted adenovirus vectors

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19990903

AK Designated contracting states

Kind code of ref document: A1

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

RIC1 Information provided on ipc code assigned before grant

Free format text: 7C 12N 5/00 A, 7C 12N 15/63 B, 7C 12N 1/21 B, 7C 12N 15/64 B, 7C 12N 5/10 B, 7C 12N 5/22 B, 7C 12N 15/86 B

A4 Supplementary search report drawn up and despatched

Effective date: 20000303

AK Designated contracting states

Kind code of ref document: A4

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

17Q First examination report despatched

Effective date: 20001215

GRAH Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOS IGRA

TPAD Observations by third parties

Free format text: ORIGINAL CODE: EPIDOS TIPA

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

Owner name: GENSTAR THERAPEUTICS CORPORATION

RIN1 Information on inventor provided before grant (corrected)

Inventor name: ZHANG, WEI-WEI

Inventor name: ALEMANY, RAMON

Inventor name: AYARES, DAVID

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

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

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

18D Application deemed to be withdrawn

Effective date: 20050210