WO1997000326A1 - Packaging systems for human recombinant adenovirus to be used in gene therapy - Google Patents

Packaging systems for human recombinant adenovirus to be used in gene therapy Download PDF

Info

Publication number
WO1997000326A1
WO1997000326A1 PCT/NL1996/000244 NL9600244W WO9700326A1 WO 1997000326 A1 WO1997000326 A1 WO 1997000326A1 NL 9600244 W NL9600244 W NL 9600244W WO 9700326 A1 WO9700326 A1 WO 9700326A1
Authority
WO
WIPO (PCT)
Prior art keywords
adenovirus
nucleic acid
cell
dna
cells
Prior art date
Application number
PCT/NL1996/000244
Other languages
French (fr)
Inventor
Frits Jacobus Fallaux
Robert Cornelis Hoeben
Abraham Bout
Domenico Valerio
Alex Jan Van Der Eb
Original Assignee
Introgene B.V.
Rijksuniversiteit Leiden
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26139401&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO1997000326(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to AU60182/96A priority Critical patent/AU731767B2/en
Priority to AT96917735T priority patent/ATE278794T1/en
Priority to IL16040696A priority patent/IL160406A0/en
Priority to DK04101716.1T priority patent/DK1445322T4/en
Priority to DE69633565T priority patent/DE69633565T3/en
Priority to EP96917735A priority patent/EP0833934B2/en
Priority to SI9630698T priority patent/SI0833934T2/en
Priority to CA2222140A priority patent/CA2222140C/en
Priority to KR10-2004-7010409A priority patent/KR100470180B1/en
Priority to US08/793,170 priority patent/US5994128A/en
Priority to IL12261496A priority patent/IL122614A0/en
Application filed by Introgene B.V., Rijksuniversiteit Leiden filed Critical Introgene B.V.
Priority to JP50294897A priority patent/JP4051416B2/en
Priority to ES96917735T priority patent/ES2231813T5/en
Priority to DK96917735.1T priority patent/DK0833934T4/en
Publication of WO1997000326A1 publication Critical patent/WO1997000326A1/en
Priority to US09/333,820 priority patent/US6306652B1/en
Priority to US09/918,029 priority patent/US6783980B2/en
Priority to US10/038,271 priority patent/US20020151032A1/en
Priority to US10/125,751 priority patent/US7105346B2/en
Priority to US10/219,414 priority patent/US20030104626A1/en
Priority to US10/618,526 priority patent/US20050260596A1/en
Priority to IL160406A priority patent/IL160406A/en
Priority to US10/850,140 priority patent/US7052881B2/en
Priority to US11/134,674 priority patent/US8236293B2/en
Priority to US11/485,114 priority patent/US20060246569A1/en
Priority to US11/879,421 priority patent/US20090023196A1/en
Priority to US11/900,463 priority patent/US20080138901A1/en

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N2710/10321Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • 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
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/15Vector systems having a special element relevant for transcription chimeric enhancer/promoter combination
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/42Vector systems having a special element relevant for transcription being an intron or intervening sequence for splicing and/or stability of RNA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/60Vector systems having a special element relevant for transcription from viruses

Definitions

  • the invention relates to the field of recombinant DNA tecnnology, more in particular to the field of gene therapy.
  • the invention relates to gene therapy using materials derived from adenovirus, in particular human recombinant adenovirus It especially relates to novel virus derived vectors and novel packaging cell lines for vectors based on adenoviruses.
  • Gene therapy is a recently developed concept for which a wide range of applications can be and have been envisaged.
  • genetic information is introduced into some or all cells of a host, as a result of which the genetic information is added to the host in a functional format.
  • the genetic information added may be a gene or a derivative of a gene, sucn as a cDNA, which encodes a protein.
  • the functional format means that the protein can be expressed by the machinery of the host cell.
  • the genetic information can also be a sequence of nucleotides complementary to a sequence of nucleotides (be it DNA or RNA) present in the host cell.
  • the functional format m this case is that the added DNA (nucleic acid) molecule or copies made thereof in situ are capable of base pairing with the complementary sequence present in the host cell.
  • Applications include the treatment of genetic disorders by supplementing a protein or other substance which is, through said genetic disorder, not present or at least present in insufficient amounts in the host, the treatment of tumors and (other) acquired diseases such as (auto) immune diseases or infections, etc.
  • tnere are basically three different approaches in gene therapy, one directed towards compensating a deficiency present in a (mammalian) host, the second directed towards the removal or elimination of unwanted substances (organisms or cells) and the third towards application of a recombinant vaccine (tumors or foreign micro-organisms) .
  • Adenoviruses are non-enveloped DNA viruses
  • Gene-transfer vectors derived from adenoviruses (so-called adenoviral vectors) have a number of features that make them
  • the biology of the adenoviruses is characterized in detail, the adenovirus is not associated with severe human pathology, the virus is extremely efficient in introducing its DNA into the host cell, the virus can infect a wioe variety of cells and has a broad host-range, the virus can be produced in large quantities with relative ease and the virus can be rendered replication defective by deletions in the earlyregion 1 (E1) of the viral genome.
  • E1 earlyregion 1
  • the adenovirus genome is a linear double-stranded DNA molecule or approximately 36000 base pairs with the 55-kDa terminal protein covalently bound to the 5' terminus of each strand
  • the Ad DNA contains identical Inverted Terminal
  • ITR Inticle Repeats
  • the viral origins of replication are located within the ITRs exactly at the genome ends DNA synthesis occurs in two stages. First, the replication proceeds by strand displacement, generating a daughter duple molecule and a parental displaced strand. The displaced strand is single stranded and can form a so-called
  • panhandle which allows replication initiation and generation of a daughter duplex molecule.
  • replication may proceed from both ends of the genome
  • the viral genes are expressed in two phases, the early p.iase, which is the period upto viral DNA replication, and the late phase, which coincides with the initiation of viral DNA replication.
  • the early phase only the early gene products, encoded by regions E1, E2, E3 and E4, are expressed, which carry out a number of functions that prepare the cell for synthesis of viral structural proteins (Berk, 1986).
  • the late phase t.ie late viral gene products are expressed in addition to the early gene products and host cell DNA and protein synthesis are shut off Consequently, the cell becomes dedicated to the production of viral DNA and of viral structural proteins (Tooze, 1981).
  • the E1 region of adenovirus is the first region of adenovirus expressed after infection of the target cell This region consists of two transcriptional units, the E1A and E1B genes, which both are required for oncogenic transformation of primary (embryonal) rodent cultures. The main functions of the E1A gene products are.
  • E1B gene ii) to transcriptionally activate the E1B gene and the other early regions (E2, E3, E4)
  • Transfection of primary cells with the E1A gene alone can induce unlimited proliferation (immortalization), but does not result in complete transformation.
  • expression cf E1A in most cases results in induction of programmed cell death (apoptosis), and only occasionally immortalization is obtained (Jochemsen et al., 1987) .
  • Co-expression of the E1B gene is required to prevent induction of apoptosis and for complete morphological transformation to occur.
  • high level expression of E1A can cause complete transformation in the absence of E1B (Roberts et al., 1985).
  • the E1B encoded proteins assist E1A in redirecting the cellular functions to allow viral replication.
  • the E1B 55 kD and E4 33kD proteins, which form a complex that is essentially localized in the nucleus, function in
  • E1B 21 kD protein is important for correct temporal control of the productive infection cycle, thereby preventing premature death of the host cell before the virus life cycle has been completed
  • Mutant viruses incapable of expressing the E1B 21 kD gene-product exhibit a shortened infection cycle that is accompanied by excessive
  • deg-phenotype degradation of host cell chromosomal DNA
  • cyt-phenotype enhanced cytopathic effect
  • the deg and cyt pnenotypes are suppressed when in addition the E1A gene is mutated, indicating that these pnenotypes are a function of E1A
  • E1B 21 kDa protein slows down the rate by wmch E1A switches on the other viral genes It is not yet known through which mechanisms) E1B 21 kD quenches these E1A dependent functions.
  • Vectors derived from human adenoviruses in which at least the E1 region has oeen deleted and replaced by a gene of interest, have oeen used extensively for gene therapy experiments in tne pre-clinical and clinical phase.
  • adenoviruses a) do not integrate into the host cell genome; b) are able to infect non-dividing cells and c) are able to efficiently transfer recombinant genes in vivo (Brody and Crystal, 1994). Those features make
  • adenoviruses attractive candidates for in vivo gene transfer of, for instance, suicide or cytokine genes into tumor cells.
  • recombinant adenovirus technology is the possibility of unwanted generation of replication competent adenovirus (RCA) during the production of recombinant adenovirus (Lochmuller et al., 1994, Imler et al., 1996) This is caused by homologous recombination between overlapping sequences from the recombinant vector and the adenovirus constructs present in the complementing cell line, such as the 293 cells (Granam et al., 1977).
  • RCA replication competent adenovirus
  • RCA in batches to be used in clinical trials is unwanted because RCA i) will replicate in an uncontrolled fashion; ii) can complement replication defective recombinant adenovirus, causing uncontrolled multiplication of the recombinant adenovirus and iii) batches containing RCA induce significant tissue damage and hence strong pathological side effects
  • this problem in virus production is solved in that we have developed packaging cells that have no overlapping sequences with a new basic vector and thus are suited for safe large scale production of recombinant adenoviruses one of the
  • adenoviruses are deleted for the E1 region (see above) .
  • the adenovirus E1 products trigger the transcription of the other early genes (E2, E3, E4 ), which consequently activate expression of the late virus genes. Therefore, it was generally thought that E1 deleted vectors would not express any other adenovirus genes.
  • the E2A protein may induce an immune response by itself and it plays a pivotal role in the switch to the synthesis of late adenovirus proteins. Therefore, it is attractive to rnaxe recombinant adenoviruses wnich are mutated m the E2 region, rendering it temperature sensitive (ts), as has been claimed in patent application WO/28938.
  • adenoviruses have been tested, and prolonged recombinant gene expression was reported (Yang et al., 1994b;
  • revertants are either real revertants or the result of second site mutations (Kruijer et al., 1983, Nicolas et al., 1981). Both types of revertants have an E2A protein that functions at normal temperature and have therefore similar toxicity as the wild-type virus.
  • E2A coding sequences from tne recombinant adenovirus genome and transfect these E2A sequences into the (packaging) cell lines containing E1 sequences to complement recombinant adenovirus vectors.
  • Major hurdles in this approach are a) that E2A should be expressed to very high levels and b) that E2A protein is very toxic to cells.
  • the current invention in yet another aspect therefore discloses use of the ts125 mutant E2A gene, which produces a protein that is not anle to bind DNA sequences at the non permissive temperature. High levels of this protein may be maintained in the cells (because it is not toxic at this temperature) until tne switch to the permissive temperature is made.
  • This can be combined witn placing the mutant E2A gene under the direction of an mducible promoter, such as for instance tet, methallothionem, steroid mducible promoter retmoic acid ⁇ -receptor or otner mducible systems.
  • an mducible promoter such as for instance tet, methallothionem, steroid mducible promoter retmoic acid ⁇ -receptor or otner mducible systems.
  • an mducible promoter such as for instance tet, methallothionem, steroid mducible promoter retmoi
  • E2A-deleted recombinant adenovirus Two salient additional advantages of E2A-deleted recombinant adenovirus are the increased capacity to harpor heterologous sequences and the permanent selection for cells that express tne mutant E2A.
  • This second advantage relates to the high frequency of reversion of ts125 mutation when reversion occurs in a cell line narooring ts125 E2A, this wi ll be lethal to tne cell.
  • E1 sequences to allow replication of E1/E2 defective adenoviruses
  • E2A sequences containing the hr mutation and the ts 125 mutation, named ts400 (Brough et al., 1985; Rice and Klessig, 1985) to prevent cell death by E2A overexpression, and/or
  • E2A sequences just containing the hr mutation, under the control of an inducible promoter, and/or
  • E2A sequences containing the hr mutation and the ts 125 mutation (ts400), under the control of an inducible promoter
  • HER human embryonic retinoblasts
  • This cell line named 911, deposited under no 95062101 at the ECACC, has many characteristics that make it superior to the commonly used 293 cells (Fallaux et al., 1996). 2. novel packaging cell lines that express just E1A genes and not E1B genes.
  • Established cell lines (and not human diploid cells of which 293 and 911 cells are derived) are able to express E1A to high levels without undergoing
  • apoptotic cell death as occurs in numan diploid cells that express E1A in the absence of E1B.
  • Such cell lines are able to trans-complement E1B- defective recombinant adenoviruses, because viruses mutated for E1B 21 kD protein are apie to complete viral replication even faster than wild-type
  • the constructs are transfected into the different established cell lines and are selected for high expression of E1A. This is done by operatively linking a selectable marker gene (e.g. NEO gene) directly to the E1B promoter.
  • a selectable marker gene e.g. NEO gene
  • the E1B promoter is transcriptionally activated by the E1A gene product and therefore resistance to the selective agent (e.g. G418 in the case NEO is used as the selection marker) results m direct selection for desired expression of the E1A gene
  • Packaging constructs that are mutated or deleted for E1B 21 kD, but just express the 55 kD protein.
  • E1A immortalized by expression of E1A.
  • E1B essential to prevent apoptosis induced by E1A proteins
  • E1 expressing cells are achieved by selection for focus formation (immortalization), as described for 293 cells (Graham et al., 1977) and 911 cells (Fallaux et al.,1996), that are E1-transformed human embryonic kidney (HEK) cells and human embryonic retmoblasts (HER), respectively.
  • HEK human embryonic kidney
  • HER human embryonic retmoblasts
  • PER.C1B After transfection of HER cells with construct pIG. E1B (Fig. 4), seven independent cell lines cculd be established. These cell lines were designated PER.C1, PER.C3, PER.C4, PER.C5, PER.C6, PER.C8 ar.d PER.C9. PER denotes PGK-E1-Retmoblasts. These cell lines express E1A and E1B proteins, are stable (e.g. PER.C6 for more than 57 passages) and complement E1 defective
  • adenovirus vectors Yields of recombinant adenovirus obtained on PER cells are a little higher than obtained on 293 cells.
  • Yields of recombinant adenovirus obtained on PER cells are a little higher than obtained on 293 cells.
  • adenoviral vectors contain pIX promoter sequences and the pIX gene, as pIX (from its natural promoter sequences) can only b expressed from the vector and not by packaging cells (l.atsui et al., 1986, Hoeben and Fallaux, pers. comm.; Imler et al., 1996).
  • E2A expressing packaging cell lines preferably based on either E1A expressing established cell lines or E1A - E1B expressing diploid cells (see unoer 2 - 4). E2A expression is either under the control of an inducible promoter or the E2A ts125 mutant is driven by either an mducible or a constitutive promoter.
  • Adenovirus packaging cells from monkey origin that are able to trans-complement E1-defective recombinant adenoviruses. They are preferably co-transfected with pIG.E1AE1B and pIG.NEO, and selected for NEO
  • adenoviruses that harbor a host-range mutation in the E2A gene, allowing human adenoviruses to replicate in monkey cells.
  • Such viruses are generated as described in Figure 12,-except DNA from a hr-mutant is used for homologous recombination.
  • Adenovirus packaging cells from monkey origin as
  • E2A in these cell lines is either under the control of an mducible promoter or the tsE2A mutant is used. In the latter case, the E2A gene will thus carry both the ts mutation and the hr mutation (derived from ts400).
  • Replication competent human adenoviruses have been described that harbor both mutations (Brough et al., 1985; Rice and Klessig, 1985).
  • a further aspect of the invention provides otherwise improved adenovirus vectors, as well as novel strategies for generation and application of such vectors and a method for the mtracellular amplification of linear DNA fragments in mammalian cells.
  • the so-called "minimal" adenovirus vectors according to the present invention retain at least a portion of the viral genome that is required for encapsidation of tne genome into virus particles (the encapsidation signal), as well as at least one copy of at least a functional part or a derivative of the Inverted Terminal Repeat (ITR), that is DNA sequences derived from the termini of the linear adenovirus genome.
  • the vectors according to the present invention will also contain a transgene linked to a promoter sequence to govern expression of the transgene.
  • Packaging of the so-called minimal adenovirus vector can be achieved by co-infection with a helper virus or, alternatively, with a packaging deficient replicating helper system as described below.
  • Adenovirus-derived DNA fragments that can replicate in suitable cell lines and that may serve as a packaging deficient replicating helper system are generated as follows. These DNA fragments retain at least a portion of the transcribed region of the "late" transcription unit of the adenovirus genome and carry deletions in at least a portion of the E1 region and deletions in at least a portion of the encapsidation signal In addition, these DNA fragments contain at least one copy of an inverted terminal repeat (ITR). At one terminus of the transfected DNA molecule an ITR is located The other end may contain an ITR, or alternatively, a DNA sequence that is
  • the free 3'-hydroxyl group of the 3' terminal nucleotide of the hairpm- structure can serve as a primer for DNA synthesis by cellular and/or adenovirus-encoded DNA polymerases, resulting in conversion into a double-stranded form of at least a portion of the DNA molecule Further replication initiating at the ITR will result in a linear double- stranded DNA molecule, that is flanked by two ITR's, and is larger than the original transfected DNA molecule (see Fig. 13).
  • This molecule can replicate itself in the transfected cell by virtue of the adenovirus proteins encoded by the DNA molecule and the adenoviral and cellular proteins encoded by genes in the host-cell genome This DNA molecule can not be encapsidated due to its large size (greater than 39000 base pairs) or due to the absence or a functional encapsidation signal. This DNA molecule is intended to serve as a helper for the production of defective adenovirus vectors in suitable cell lines.
  • the invention also comprises a method for the amplification of linear DNA fragments of variable size in suitable mammalian cells.
  • DNA fragments contain at least one copy of the ITR at one of the termini of the fragment.
  • the other end may contain an ITR, or
  • a DNA sequence that is complementary to a portion of the same strand of the DNA molecule other than the ITR If, in the latter case, the two complementary sequences anneal, the free 3'-hydroxyl group of the
  • 3' terminal nucleotide of the hairpin-structure can serve as a primer for DNA synthesis by cellular and/or
  • a DNA molecule that contains ITR sequences at both ends can replicate itself in transfected cells by virtue of the presence of at least the adenovirus E2 proteins (viz. the DNA-bmding protein (DBP), the
  • Ad-pol adenovirus DNA polymerase
  • pTP preterminal protein
  • adenovirus E2 genes integrated in the host-cell genome, or from a replicating helper fragment as described above.
  • defective molecules with a total length of up to two times the normal genome could be generated. Such molecules could contain duplicated sequences from either enc of the genome. However, no DNA molecules larger than the full- length virus were found packaged in the defective
  • Ad A The strong immunogenicity of the adenovirus particle results m an immunological response of the host, even after a single administration of the adenoviral vector. As a result of the development of neutralizing antibodies, a subsequent administration of the virus will be less effective or even completely ineffective. However, a prolonged or persistent expression of the transferred genes will reduce the number of administrations required and may bypass the problem.
  • adenovirus proteins encoded by the viral vector, were expressed despite deletion of the E1 region.
  • the E2A region can be deleted from the vector, when the E2A gene product is provided in trans in the encapsidation cell line, adding another 1.6 kb. It is, however, unlikely that the maximum capacity of foreign DNA can be significantly increased further than 12 kb.
  • helperfree-stocks of recombinant adenovirus vectors that can accomodate up to 38 kb of foreign DNA. Only two functional ITR sequences, and sequences that can function as an encapsidation signal need to be part of the vector genome. Such vectors are called minimal
  • the helper functions for the minimal adenovectors are provided in trans by encapsidation defective-replication competent DNA molecules that contain all the viral genes encoding the required gene products, with the exception of those genes that are present in the host- cell genome, or genes that reside in the vector genome.
  • the constructs in particular pIG.E1A.E1B, will be used to transfect diploid human cells, such as Human Embryonic Retinoblasts (HER), Human Embryonic Kidney cells (HEK), and Human Embryonic Lung cells (HEL). Transfected cells will be selected for transformed phenotype (focus formation) and tested for their ability to support propagation of E1-deleted recombinant adenovirus, such as IG.Ad.MLPI.TK. Such cell lines will be used for the generation and (large-scale) production of E1-deleted recombinant adenoviruses. Such cells, infected with recombinant adenovirus are also intended to be used in vivo as a local producer of recombinant adenovirus, e.g. for the treatment of solid tumors.
  • HER Human Embryonic Retinoblasts
  • HEK Human Embryonic Kidney cells
  • HEL Human Emb
  • 911 cells are used for the titration, generation and production of recombinant adenovirus vectors (Fallaux et al., 1996).
  • HER cells transfected with pIG.E1A.E1B has resulted in 7 independent clones (called PER cells). These clones are used for the production of E1 deleted (including non- overlapping adenovirus vectors) or E1 defective
  • E2B or E2A constructs e.g. ts125E2A, see below
  • E4 etc. that will allow propagation of adenovirus vectors that have mutations in e.g. E2A or E4.
  • diploid cells of otner species that are permissive for human adenovirus, such as the cotton rat
  • Such cells infected with recombinant adenovirus, are also intended to be used in vivo for the local production of recombinant adenovirus, e.g. for the treatment of solid tumors.
  • the constructs in particular pIG.E1A.NEO, can be used to transfect established cells, e.g. A549 (human bronchial carcinoma), KB (oral carcinoma), MRC-5 (human diploid lung cell line) or GLC cell lines (small cell lung cancer) (de Leij et al., 1985; Postmus et al., 1988) and selected for NEO resistance. Individual colonies of resistant cells are isolated and tested for their capacity to support propagation of E1 -deleted recombinant
  • adenovirus such as IG.Ad.MLPI.TK.
  • E1 deleted viruses on E1A containing cells can be used for the generation and production of E1-deleted recombinant adenovirus. They are also be used for the propagation of E1A deleted/E1B retained
  • Established cells can also be co-transfected with pIG.E1A.E1B and pIG.NEO (or another NEO containing expression vector)
  • Clones resistant to G418 are tested for their ability to support propagation of II deleted recombinant adenovirus, such as IG.Ad.MLPI.TK and used for the generation and production of E1 deleted recombinant adenovirus and will be applied m vivo for lccal
  • All cell lines including transformed diploid cell lines or NEO-resistant established lines, can be used as the basis for the generation of 'next generation'
  • E1-defective recombinant adenoviruses that also carry deletions in other genes, such as E2A and E4. Moreover, they will provide the basis for the generation of minimal adenovirus vectors as disclosed herein.
  • Packaging cells expressing E2A sequences are and will be used for the generation and (large scale) production of E2A-deieted recombinant adenovirus.
  • the newly generated human aoenovirus packaging cell lines or cell lines derived from species permissive for human adenovirus E2A or ts125E2A; E1A + E2A; E1A + E1B + E2A; E1A + E2A/ts125, E1A + E1B + E2A/ts125
  • non- permissive cell lines such as monkey cells (hrE2A or hr + ts125E2A; E1A + hrE2A; E1A + E1B + hrE2A; E1A +
  • hrE2A/ts125 are and will be used for the generation and (large scale) production of E2A deleted recombinant adenovirus vectors In addition, they will be applied in vivo for local production of
  • the newly developed adenovirus vectors narboring an E1 deletion of nt. 459-3510 will be used for gene transfer purposes. These vectors are also the basis for the development of further deleted adenovirus vectors that are mutated for e.g. E2A, E2B or E4. Such vectors will be generated e.g. on the newly developed packaging cell lines described above (see 1-3).
  • adenovirus packaging constructs to be used for the packaging of minimal adenovirus vectors may nave the following characteristics:
  • the packaging construct can not be packaged because the packaging signal is deleted
  • the packaging construct contains an internal hairpm- forming sequence (see section 'Experimental, suggested hairpin' see Fig. 15)
  • packaging construct is duplicated, that is the DNA of the packaging construct becomes twice as long as it was before transfection into the packaging cell (in our sample it duplicates from 35 kb to 70 kb ) .
  • This duplication also prevents packaging. Note that this duplicated DNA molecule has ITR's at both termini (see e.g. Fig. 13)
  • this duplicated packaging molecule is able to
  • the packaging construct has no overlapping sequences with the minimal vector or cellular sequences that may lead to generation of RCA by homologous
  • This packaging system will be used to produce minimal adenovirus vectors.
  • the advantages of minimal adenovirus vectors e.g. for gene therapy of vaccination purposes, are well known (accommodation of up to 38 kb; gutting of all potentially toxic and lmmunogenic adenovirus genes).
  • Adenovirus vectors containing mutations in essential genes can also be propagated using this system. Use of intracellular E2 expressing vectors.
  • Minimal adenovirus vectors are generated using the helper functions provided in trans by packaging-deficient replicating helper molecules.
  • the adenovirus-derived ITR sequences serve as origins of DNA replication in the presence of at least the E2-gene products.
  • the E2 gene products are expressed from genes in the vector genome (N.B the gene (s) must be driven by an E1- independent promoter)
  • the vector genome can replicate in the target cells. This will allow an significantly increased number of template molecules in the target cells, and, as a result an increased expression of the genes of interest encoded by the vector. This is of particular interest for approaches of gene therapy in cancer.
  • DNA fragments of known or unknown sequence could be amplified in cells containing the E2-gene products if at least one ITR sequence is located near or at its terminus.
  • ITR sequence located near or at its terminus.
  • Even fragments much larger than the adenovirus genome (36 kb) should be amplified using this approach. It is thus possible to clone large fragments in mammalian cells without either shuttling the fragment into bacteria (such as E. coli) or use the polymerase chain reaction (P.C.R.).
  • P.C.R. polymerase chain reaction
  • the linear DNA fragments can be amplif ⁇ ed to similar levels
  • This system can be used to express heterologous proteins (equivalent to the
  • Simian Virus 40-based COS-cell system ror research or for therapeutic purposes
  • the system can be used to identify genes in large fragments of DNA. Random DNA fragments may be amplified (after addition of ITRs) and expressed during mtracellular amplification. Election or, selection of those cells with the oesired pnenotvpe can be used to enrich the fragment of interest and to isolate the gene.
  • This cell line was obtained by transrection of human diploid human embryonic retinoblasts (HER) with pAd5XhoIC, that contains nt. 80 - 5788 of Ad5; one of the resulting transformants was designated 911.
  • This cell line has been shown to be very useful in the propagation of E1 defective recombinant adenovirus. It was found to be superior to the 293 cells. Unlike 293 cells, 911 cells lack a fully transformed pnenotvpe, which most likely is the cause of performing better as adenovirus packaging line:
  • plaque assays can be performed faster (4 - 5 days instead of 8-14 days on 293)
  • 911 cells were transfected using a defined construct. Transfection efficiencies of 911 cells are comparable to those of 293.
  • Adenovirus sequences are derived either from
  • pAd5. Sa1B containing nt. 80 - 9460 of human adenovirus type 5 (Bernards et al., 1983) or from wild-type Ad5 DNA.
  • pAd5. Sa1B was digested with Sall and Xhol and the large fragment was religated and this new clone was named pAd5. X/S.
  • IntrcGene The Netherlands was used as a source for the human PGK promoter and the NEO gene.
  • E1A sequences in the new packaging constructs is driven by the human PGK promoter (Michelson et al., 1983; Singer-Sam et al., 1984), derived from plasmid pTN (gift of R. Vogels), which uses pUC119 (Vieira and Messing, 1987) as a backbone.
  • This plasmid was also used as a source for NEO gene fused to the Hepatitis B Virus (HBV) poly-adenylation signal. Fusion of PGK promoter to E1 genes (Fig. 1)
  • Vector pTN was digested with restriction enzymes EcoRI (partially) and Seal, and the DNA fragment
  • PBS.PGK.PCRI contains the human PGK promoter operatively linked to Ad5 E1 sequences from nt 459 to nt. 916.
  • pIG.E1A.E1B.X Construction of pIG.E1A.E1B.X (Fig. 2) pIG.E1A.E1B.X was made by replacing the Scal-BspEI fragment of pAT-X/S by the corresponding fragment from PBS.PGK.PCRI (containing the PGK promoter linked to E1A sequences).
  • pIG.E1A.E1B.X contains the E1A and E1B coding sequences under the direction of the PGK promoter.
  • Ad5 sequences from nt.459 to nt 5788 are present in this construct, also pIX protein of adenovirus is encoded by this plasmid.
  • the Ncol - StuI fragment of pTN containing the NEO gene and part of the Hepatitis B Virus (HBV) poly-adenylation signal, was cloned into pAT-X/S-PCR2 (digested with Ncol and Nrul).
  • the poly- adenylation signal was completed by replacing the Scal- Sall fragment of pAT-PCR2-NEO by the corresponding fragment of pTN (resulting in pAT.PCR2.NEO.p(A)).
  • the Seal - Xbal of pAT.PCR2.NEO.p (A) was replaced by the
  • the resulting construct was named pIG.E1A.NEO, and thus contains Ad5 E1 sequences (nt 459 to nt 1713) under the control of the human PGK promoter.
  • pIG.EIA.E1B Construction of pIG.EIA.E1B (Fig. 4) pIG.E1A.E1B was made by amplifying the sequences encoding the N-terminal ammo acids of E1B 55kd using primers Eb1 and Eb2 (introduces a Xhol site). The
  • pIG.E1A.E1B was constructed by introducing the HBV poly(A) sequences of pIG.E1A.NEO downstream of E1B sequences of pAT-PCR3 by exchange of Xbal - Sall fragment of pig.E1A.NEO and the Xbal Xhol fragment of pAT PCR3
  • pIG.EIA.E1B contains nt. 459 to nt. 3510 of Ad5, that encode the E1A and E1B proteins.
  • the E1B sequences are terminated at the splice acceptor at nt.3511. No pIX sequences are present in this construct.
  • Construction of pIG.NEO (Fig. 5) pIG NEO was generated by cloning the Hpal - Seal fragment of pIG.E1A.NEO, containing the NEO gene under the control of the Ad.5 E1B promoter, into pBS digested with EcoRV and Seal.
  • This construct is of use when established cells are transfected with E1A.
  • E1B constructs and NEO selection is required. Because NEO expression is directed by the E1B promoter, NEO resistant cells are expected to co-express E1A, which also is advantageous for maintaining high levels of expression of E1A during long-term culture of the cells. Testing of constructs.
  • pIG.E1A.E1B.X and pIG.E1A.E1B was assessed by restriction enzyme mapping, furthermore, parts of the constructs that were obtained by PCR analysis were confirmed by sequence analysis No changes in the nucleotide sequence were found.
  • constructs were transfected into primary BRK (Baby Rat Kidney) cells and tested for their ability to immortalize (pIG.E1A.NEO) or fully transform
  • Kidneys of 6-day old WAG-Rij rats were isolated, homogenized and trvpsmized.
  • Subconfluent disnes (diameter 5 cm) of the BRK cell cultures were transfected with 1 or 5 ⁇ g of pIG.NEO, pIG.E1A.NEO, pIG.E1A.E1B, pIG.E1A.E1B.X, pAd5XhoIC, or with pIG.E1A.NEO together with PDC26
  • HER cells were grown in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% Fetal Calf Serum (FCS) and antibiotics in a 5% CO2 atmospnere at 37°C.
  • DMEM Dulbecco's Modified Eagle Medium
  • FCS Fetal Calf Serum
  • FCS Fetal Calf Serum
  • Cell culture media, reagents and sera were purchased from Gibeo Laboratories (Grand Island, NY). Culture plastics were purchased from Gremer (Nurtmgen, Germany) and Corning (Corning, NY).
  • the recombinant adenoviral vector IG.Ad.MLP.nls.lacZ contains the E.coli lacZ gene, encoding ⁇ -gaiactosidase, under control of the Ad2 major late promoter
  • MLP IG.Ad.MLP.luc
  • IG.Ad.MLP.TK contains the firefly luciferase gene driven by the Ad2 MLP Adenoviral vectors IG.Ad.MLP.TK and IG.Ad.CMV.TK contain the Herpes Simplex Virus thymidine kmase (TK) gene under the control of the Ad2 MLP and the Cytomegalovirus (CMV) enhancer/promoter, respectively.
  • Ad5-E1-transformed A549 and PER cells were cashed with PBS and scraped in Fos-RIPA buffer (10 mM Tris (pH 7,5), 150 mM NaCl, 1% NP40,01% sodium dodecyl sulphate (SDS), 1% NA-DOC, 0,5 mM phenyl methyl sulphonyl fluoride PMSF), 0,5 mM trypsin inhibitor, 50 mM NaF and 1 mM sodium vanadate). After 10 min. at room temperature, lysates were cleared by centrifugation. Protein
  • First antibodies were the mouse monoclonal anti-Ad5-E1B- 55-kDA antibody A1C6 (Zantema et al., unpublished), the rat monoclonal anti-Ad5-E1B-221-kDa antibody ClGll (Zantema et al., 1985).
  • the second antibody was a
  • Ad5-E1-transformed A549 human bronchial carcinoma cell lines were generated by transfection with pIG.E1A.NEO and selection for G418 resistance. Thirty-one G418 resistant clones were established. Co-transfection of pIG.EIA.E1B with pIG.NEO yielded seven G418 resistant cell lines.
  • Ad5-E1-transformed human embryonic retina (HER) cells were generated by transfection of primery HER cells with plasmid pIG.EIA.E1B. Transformed cell lines were
  • PER.C1 PER.C3, PER.C4, PER.C5, PER.C6, PER.C8 and PER.C9.
  • One of the PER clones namely PER.C6, has been deposited at the ECACC under number 96022940.
  • Ad5 E1A and the 55-kDa and 21 kDa E1B proteins in the established A549 and PER cells was studied by means of Western blotting, with the use of monoclonal antibodies (mAb).
  • mAb monoclonal antibodies
  • Mab M73 recognizes the E1A products, whereas Mabls AIC6 and ClGll are directed against the 55 -kDa and 21 kDa E1B proteins, respectively.
  • the antibodies did not recognize proteins in extracts from the parental A549 or the primary HER cells (data not shown). None of the A549 clones that were generated by co-transfection of pIG.NEO and pIG.E1A.E1B expressed detectable levels of E1A or E1B proteins (not shown). Some of the A549 clones that were generated by transfection with pIG.E1A.NEO expressed the Ad5 E1A proteins (Fig. 7), but the levels were much lower than those detected in protein lysates from 293 cells. The steady state E1A levels detected in protein extracts from PER cells were much higher than those detected m extracts from A549- derived cells All PER cell lines expressed similar levels of E1A proteins (Fig. 7).
  • E1B proteins particularly in the case of E1B 55 kDa, was mote variable. Compared to 911 and 293, the majority of the PER clones express high levels of E1B 55 kDa and 21 kDa. The steady state level of E1B 21 kDa was the highest in
  • PER. C1, C3, C4, C5, C6, C8 and C9 contain 2, 88, 5,4, 5, 5 and 3 copies of the Ad5 E1 coding region, respectively, and that 911 and 293 cells contain 1 and 4 copies of the Ad5 E1 sequences, respectively.
  • Recombinant adenovectors are generated by cotransfection of adaptor plasmids and the large Clal fragment of Ad5 into 293 cells (gee patent application EP 95202213).
  • the recombinant virus DNA is formed by homologous recombination between the homologous viral sequences that are present in the plasmid and the
  • the yields of the novel adenovirus vector IG.Ad.MLPI.TK are similar or higher than the yields obtained for the other viral vectors on all cell lines tested.
  • the used recombinant adenovirus vectors are deleted for E1 sequences from 459 to nt. 3328.
  • E1B contains Ad5 sequences 459 to nt. 3510 there is a sequence overlap of 183 nt. between E1B sequences in the packaging construct pIG.E1A.E1B and recombinant adenoviruses, sucn as e.g. IG.Ad.MLP.TK.
  • the overlapping sequences were deleted from the new adenovirus vectors.
  • non-coding sequences derived from lacZ, that are present in the original contructs were deleted as well. This was achieved (see Fig.
  • SV40 poly (A) sequences from pMLP.TK using primers SV40-1 (introduces a BamHI site) and SV40-2 (introduces a Bglll site).
  • Ad5 sequences present in this construct were amplified from nt 2496 (Ad5, introduces a Bglll site) to nt. 2779 (Ad5-2). Both PCR fragments were digested with Bglll and were ligated. The ligation product was PCR amplified using primers SV40-1 and Ad5-2. The PCR product obtained was cut with BamHI and Aflll and was ligated into pMLP.TK predigested with the same enzymes.
  • the resulting construct named pMLPI.TK, contains a deletion in adenovirus E1 sequences from nt 459 to nt 3510.
  • pIG.E1A.NEO when transfected into established cells, is expected to be sufficient to support propagation of E1-deleted recombinant adenovirus. This combination does not have any sequence overlap, preventing generation of RCA by homologous recombination.
  • this convenient packaging system allows the propagation of recombinant adenoviruses that are deleted just for E1A sequences and not for E1B sequences.
  • TNF Tumor Necrosis Factor
  • Recombinant adenovirus was generated by cotransfection of 293 cells with Sail linearized pMLPI.TK DNA and Clal linearized Ad5 wt DNA. The procedure is schematically represented in Fig. 12.
  • restriction endonuclease Asp718 in its correct and in the. reverse orientation, respectively (Fig. 15).
  • Eg. pICLhaw is a plasmid that contains the adenovirus ITR followed by the CMV-driven luciferase gene and the Asp718 hairpin m the reverse (non-functional)
  • Plasmids pICLhac, pICLhaw, pICLI and pICL were generated using standard techniques. The schematic representation of these plasmids is shown in Figs. 16-19.
  • Plasmid pICL is derived from the following plasmids: nt.1 - 457 pMLP10 (Levrero et al., 1991)
  • nt.458 - 1218 pCMV ⁇ (Clontech, EMBL Bank No. U02451) nt.1219 - 3016 pMLP . luc (IntroGene, unpublished) nt.3017 - 5620 pBLCAT5 (Stem and Whelan, 1989)
  • the plasmid has been constructed as follows:
  • the tet gene of plasmid pMLP10 has been inactivated by deletion of the BamHI-Sail fragment, to generate pMLP10 ⁇ SB.
  • primer set PCR/MLP1 and PCR/MLP3 a 210 bp fragment containing the Ad5-ITR, flanked by a synthetic Sall restriction site was amplified using pMLP10 DNA as the template.
  • the PCR product was digested with the enzymes EcoRI and SgrAI to generate a 196 bp. fragment.
  • Plasmid pMLP10 ⁇ SB was digested with EcoRI and SgrAI to remove the ITR. This fragment was replaced by the EcoRI- SgrAI-treated PCR fragment to generate pMLP/SAL .
  • Plasmid pCMV-Luc was digested with PvuII to completion and recirculated to remove the SV40-derived poly-adenylation signal and Ad5 sequences with exception of the Ad5
  • Ad5 ITR was replaced by the Sal-site-flanked ITR from plasmid pMLP/SAL by exchanging the Xmnl-SacII fragments.
  • the resulting plasmid, pCMV-luc ⁇ Ad/SAL, the Ad5 left terminus and the CMV-driven luciferase gene were isolated as an Sall-Smal fragment and inserted in the Sall and Hpal digested plasmid pBLCATS, to form plasmid pICL.
  • Plasmid pICL is represented in Fig 19; its sequence is presented in Fig. 20.
  • Plasmid pICL contains the following features nt. 1-457 Ad5 left terminus (Sequence 1-457 of
  • nt. 1218-2987 Firefly luciferase gene (from pMLP.luc) nt. 3018-3131 SV40 tandem poly-adenylation signals from late transcript, derived from piasmid pBLCAT5)
  • pUC12 backbone (derived from plasmid
  • Plasmids pICLhac and pICLhaw were derived from plasmid pICL by digestion of the latter plasmid with the restriction enzyme Asp718
  • the linearized plasmid was treated with Calf-Intestine Alkaline Phosphatase to remove the 51 phoshate groups.
  • the partially complementary synthetic single-stranded oligonucleotide Hp/aspi en Hp/asp2 were annealed and phosphorylated on their 5' ends using T4-polynucleot ⁇ de kmase.
  • the phosporylated double-stranded oligomers were mixed with the dephosporylated pICL fragment and ligated. Clones containing a single copy of the synthetic oligonucleotide inserted into the plasmid were isolated and characterized using restriction enzyme digests. Insertion of the oligonucleotide into the Asp718 site will at one junction recreate an Asp718 recognition site, whereas at the other junction the recognitionsite will be disrupted. The orientation and the integrity of the inserted
  • oligonucleotide was verified in selected clones by sequence analyses.
  • a clone containing the oligonucleotide in the correct orientation was denoted pICLhac.
  • a clone with the oligonucleotide in the reverse orientation was designated pICLhaw.
  • Plasmids pICLhac and pICLhaw are represented in Figs 16 and 17.
  • Plasmid pICLI was created from plasmid pICL by insertion of the Sall-SgrAI fragment rrom pICL, containing the Ad5-ITR into the Asp718 site of pICL.
  • the 194 bp Sall- SgrAI fragment was isolated from pICL, and the cohesive ends were converted to blunt ends using E.coli DNA polymerase I (Klenow fragment) and dNTP's.
  • the Asp718 cohesive ends were converted to blunt ends by treatment with mungbean nuclease.
  • By ligation clones were generated that contain the ITR in the Asp718 site of plasmid pICL.
  • a clone that contained the ITR fragment in the correct orientation was designated pICLI (Fig. 18).
  • adenovirus was constructed according to the method described in Patent application 95202213. Two components are required to generate a recombinant adenovirus. First, an adaptor-plasmid containing the left terminus of the adenovirus genome containing the ITR and the packaging signal, an expression cassette with the gene of interest, and a portion of the adenovirus genome which can be used for homologous recombination. In addition, adenovirus DNA is needed for recombination with the aforementioned adaptor plasmid In the case of Ad-CMV-hcTK, the plasmid PCMV.TK was used as a basis This plasmid contains nt 1-455 of the adenovirus type 5 genome, nt.
  • piasmid pCMV.TK was digested with Clal (the unique Clal-site is located just upstream of the TK open readmgframe) and dephosphorylated with Calf-Intestine Alkaline Phosphate.
  • Clal the unique Clal-site is located just upstream of the TK open readmgframe
  • Calf-Intestine Alkaline Phosphate the unique Clal-site is located just upstream of the TK open readmgframe
  • oligonucleotides HP/cla2 and HP/cla2 were annealed and phopsphorylated on their 5'-OH groups with T4- polynucleotide kinase and ATP.
  • the double-stranded oligonucleotide was ligated with the linearized vector fragment and used to transform E. coli strain "Sure".
  • oligonucleotide Insetion of the oligonucleotide into the Clal site will disrupt the Clal recognition sites. In the oligonucleotide contains a new Clal site near one of its termini. In selected clones, the orientation and the megrity of the inserted oligonucleotide was verified by sequence
  • a clone containing the oligonucleotide in the correct orientation was denoted pAd-CMV-hcTK.
  • This plasmid was co-transfected with Clal digested wild-type Adenovirus-typeS DNA into 911 cells.
  • a recombinant adenovirus in wnich the CMV-hcTK expression cassette replaces the E1 sequences was isolated and propagated using standard procedures.
  • the plasmid pICLhac is introduced into 911 cells (human embryonic retinoblasts transformed with the adenovirus E1 region).
  • the plasmid pICLhaw serves as a control, which contains the
  • plasmids pICLI and pICL In the plasmid pICLI the hairpin is replaced by an adenovirus ITR Plasmid pICL contains neither a hairpin nor an ITR sequence. These plasmids serve as controls to determine the efficiency of replication by virtue of the terminal-hairpin structure.
  • the cultures are infected with the virus IG.Ad.MLPI.TK after transfection.
  • aforementioned plasmids and infected with IG.Ad.MLPI.TK virus is being analyzed by Southern blotting for the presence of the expected replication intermediates, as well as for the presence of the duplicated genomes.
  • virus is isolated, that is capable to transfer and express a luciferase marker gene into luciferase negative cells.
  • Plasmid DNA of plasmids pICLhac, pICLhaw, pICLI and pICL have been digested with restriction endonuclease Sall and treated with mungbean nuclease to remove the 4 nucleotide single-stranded extension of the resulting DNA fragment. In this manner a natural adenovirus 5' ITR terminus on the DNA fragment is created. Subsequently, both tne pICLhac and pICLhaw plasmids were digested with restriction endonuclease Asp718 to generate tne terminus capable of forming a hairpin structure. The digested plasmids are introduced into 911 cells, using the standard calcium phosphate co-precipitation technique, four dishes for each plasmid. During the transfection, for each plasmid two of the cultures are infected with the
  • IG.Ad.MLPI.TK virus using 5 infectious IG.Ad.MLPI.TK particles per cell.
  • Ad Ad.
  • tk-virus-mfected and one unmfected culture are used to isolate small cells
  • a hybridizing fragment of approx. 2.6kb is detected only in the samples from the adenovirus infected cells transfected with plasmid pICLhac.
  • the size of this fragment is consistent with the anticipated duplication of the luciferase marker gene. This supports the conclusions that the inserted hairpin is capable to serve as a primer for reverse strand synthesis.
  • the hybridizing fragment is absent if the IG.Ad.MLPI.TK virus is omitted, or if the hairpin oligonucleotide has been inserted in the reverse orientation.
  • restriction endonuclease Dpnl recognizes the tetranucleotide sequence 5'-GATC-3', but cleaves only methylated DNA, (that is, only (plasmid) DNA propagated in, and derived, from E.coli, not DNA that has been replicated m mammalian cells).
  • the restriction endonuclease Dpnl recognizes the tetranucleotide sequence 5'-GATC-3', but cleaves only methylated DNA, (that is, only (plasmid) DNA propagated in, and derived, from E.coli, not DNA that has been replicated m mammalian cells).
  • endonuclease Mbol recognizes the same sequences, but cleaves only unmethylated DNA (viz. DNA propagated m mammalian cells). DNA samples isolated from the
  • transfected cells are incubated with Mbol and Dpnl and analysed with Southern blots. These results demonstrate that only in the cells transfected with the pICLhac and the pICLI plasmids large Dpnl-resistant fragments are present, that are absent in the Mbol treated samples. These data demonstrate that only after transfection of plasmids pICLI and pICLhac replication and duplication of the fragments occur.
  • sequence that is capable of forming a hairpin structure, are sufficient to generate DNA molecules that can be encapsidated into virions.
  • virus containing two copies of the CMV-luc marker gene can be encapsidated into virions, virus is harvested from the remaining two cultures via three cycles of freeze-thaw crushing and is used to infect murme fibroblasts. Fortyeight hours after infection the infected cells are assayed for luciferase activity. To exclude the possibility that the luciferase activity has been induced by transfer of free DNA, rather than via virus particles, virus stocks are treated with DNasel to remove DNA contaminants
  • luciferase activity is only found in the cells after infection with the virus stocks derived from IG.Ad.MLPI.TK-infected cells
  • ITR at least part of the encapsidation signal as well as a synthetic DNA sequence, that is capable of forming a hairpin structure, have the intrinsic capacity to
  • the Ad-CMV-hcTK adenoviral vector has been developed. Between the CMV enhancer/promoter region and the thvmidme kmase gene the annealed oligonucleotide pair HP/cla 1 and 2 is inserted.
  • the vector Ad-CMV-hcTK can be propagated and produced in 911 cell using standard procedures. This vector is grown and propagated
  • DNA of the adenovirus Ad-CMV-hcTK is isolated from virus particles that had been purified using CsCl density- gradient centnfugation by standard techniques.
  • the virus DNA has been digested with restriction endonuclease Clal.
  • the digested DNA is s ize- fractionated on an 0.7% agarose gel and the large fragment is isolated and used for further experiments.
  • Cultures of 911 cells are transfected large Clal-fragment of the Ad-CMV-hcTK DNA using the standard calcium phosphate co-precipitation technique.
  • the AD-CMV-hc will replicate starting at the right-hand ITR.
  • a hairpin can be formed at the left-hand terminus of the fragment. This facilitates the DNA polymerase to elongate the chain towards the rignt-hand-side. The process will proceed until the displaced strand is completely converted to its double-stranded form. Finally, the right-nand ITR will be recreated, and in this location the normal adenovirus replication-initiation and elongation will occur. Note that the polymerase will read through the hairpin, thereby duplicating the molecule.
  • the resulting DNA molecule will consist of a palindromic structure of approximately 66500 bp.
  • This structure can be detected m low-molecular weight DNA extracted from the transfected cells using Southern analysis.
  • the palindromic nature of the DNA fragment can be demonstrated by digestion of the low- molecular weight DNA with suitable restriction
  • the HSV-TK gene As the probe.
  • This molecule can replicate itself in the transfected cells by virtue of the aoenovirus gene products that are present in the cells.
  • the adenovirus genes are expressed from templates that are integrated in the genome of the target cells (viz. the E1 gene products), the other genes reside in the replicating DNA fragment itself. Note however, that this linear DNA fragment cannot be encapsidated into virions. Not only does it lack all the DNA sequences required for
  • WO 94/12649 Another approach for the generation of minimal adenovirus vectors has been disclosed in WO 94/12649.
  • the method described in WO 94/12649 exploits the function of the protein IX for the packaging of minimal adenovirus vectors (Pseudo Adenoviral Vectors (PAV) in the terminology of WO 94/12649).
  • PAVs are produced by cloning an expression plasmid with the gene of interest between the left-hand (including the sequences required for encapsidation) and the right-hand adenoviral ITRs. The PAV is propagated in the presence of a helper virus.
  • Encapsidation of the PAV is preferred compared the helper virus because the helper virus is partially defective for packaging.
  • the authors propose that in the absence of the protein IX gene the PAV will be preferentially packaged.
  • neither of these mechanisms appear to be sufficiently restrictive to allow packaging of only PAVs/mmimal vectors.
  • the mutations proposed in the packaging signal dimmish packaging, but do not provide an absolute block as the same packagingactivity is required to propagate the helper virus
  • neither an increase in the size of the helper virus nor the mutation of the protein IX gene will ensure that PAV is packaged exclusively.
  • the method described in WO 94/12649 is unlikely to be useful for the production of helper-free stocks of minimal adenovirus vectors/PAVs.
  • DNA-bmdmg protein that lead to extended host range or temperature-sensitive phenotypes. J . Virol . 55, 206-212.
  • region E1B of human adenoviruses in tne absence of region E1A is not sufficient for complete transformation.
  • packaging domain is composed of a repeated element that is functionally redundant. J. Virol . 64,
  • Novel complementation cell lines derived from human lung carcinoma A549 cells support the growth of E1 -deleted adenovirus vectors.
  • adenovirus H5ts125 and H5ts107 mutants m the DNA binding protein: isolation of a new class of host range temperature conditional revertants. Virology 108, 521-524.
  • Insulin gene enhancer activity is inhibited by adenovirus 5 E1A gene

Abstract

The invention provides improved methods and products based on adenoviral materials which can advantageously be used in for instance gene therapy. In one aspect an adenoviral vector is provided which has no overlap with a suitable packaging cell line which is another aspect of invention. This combination excludes the possibility of homologous recombination, thereby excluding the possibility of the formation of replication competent adenovirus. In another aspect an adenovirus based helper construct which by its size is incapable of being encapsidated. This helper virus can be transferred into any suitable host cell making it a packaging cell. Further a number of useful mutations to adenoviral based materials and combinations of such mutations are disclosed, which all have in common the safety of the methods and the products, in particular avoiding the production of replication competent adenovirus and/or interference with the immune system. Further a method of intracellular amplification is provided.

Description

Title: Packaging systems for human recombinant adenovirus to be used in gene therapy.
The invention relates to the field of recombinant DNA tecnnology, more in particular to the field of gene therapy. In particular the invention relates to gene therapy using materials derived from adenovirus, in particular human recombinant adenovirus It especially relates to novel virus derived vectors and novel packaging cell lines for vectors based on adenoviruses.
Gene therapy is a recently developed concept for which a wide range of applications can be and have been envisaged.
In gene therapy a molecule carrying genetic
information is introduced into some or all cells of a host, as a result of which the genetic information is added to the host in a functional format.
The genetic information added may be a gene or a derivative of a gene, sucn as a cDNA, which encodes a protein. In this case the functional format means that the protein can be expressed by the machinery of the host cell.
The genetic information can also be a sequence of nucleotides complementary to a sequence of nucleotides (be it DNA or RNA) present in the host cell. The functional format m this case is that the added DNA (nucleic acid) molecule or copies made thereof in situ are capable of base pairing with the complementary sequence present in the host cell.
Applications include the treatment of genetic disorders by supplementing a protein or other substance which is, through said genetic disorder, not present or at least present in insufficient amounts in the host, the treatment of tumors and (other) acquired diseases such as (auto) immune diseases or infections, etc. As may be clear from the above, tnere are basically three different approaches in gene therapy, one directed towards compensating a deficiency present in a (mammalian) host, the second directed towards the removal or elimination of unwanted substances (organisms or cells) and the third towards application of a recombinant vaccine (tumors or foreign micro-organisms) .
For the purpose of gene therapy adenoviruses carrying deletions have been proposed as suitable. vehicles
Adenoviruses are non-enveloped DNA viruses Gene-transfer vectors derived from adenoviruses (so-called adenoviral vectors) have a number of features that make them
particularly useful for gene transfer for such purposes Eg the biology of the adenoviruses is characterized in detail, the adenovirus is not associated with severe human pathology, the virus is extremely efficient in introducing its DNA into the host cell, the virus can infect a wioe variety of cells and has a broad host-range, the virus can be produced in large quantities with relative ease and the virus can be rendered replication defective by deletions in the earlyregion 1 (E1) of the viral genome.
The adenovirus genome is a linear double-stranded DNA molecule or approximately 36000 base pairs with the 55-kDa terminal protein covalently bound to the 5' terminus of each strand The Ad DNA contains identical Inverted Terminal
Repeats (ITR) of about 100 base pairs witn the exact length depending on the serotype. The viral origins of replication are located within the ITRs exactly at the genome ends DNA synthesis occurs in two stages. First, the replication proceeds by strand displacement, generating a daughter duple molecule and a parental displaced strand. The displaced strand is single stranded and can form a so-called
"panhandle" intermediate, which allows replication initiation and generation of a daughter duplex molecule. Alternatively, replication may proceed from both ends of the genome
simultaneously, obviating the requirement to form the pannandle structure. The replication is summarized in Figure 14 adapted from (Lechner and Kelly, 1977).
During the productive infection cycle, the viral genes are expressed in two phases, the early p.iase, which is the period upto viral DNA replication, and the late phase, which coincides with the initiation of viral DNA replication. During the early phase only the early gene products, encoded by regions E1, E2, E3 and E4, are expressed, which carry out a number of functions that prepare the cell for synthesis of viral structural proteins (Berk, 1986). During the late phase t.ie late viral gene products are expressed in addition to the early gene products and host cell DNA and protein synthesis are shut off Consequently, the cell becomes dedicated to the production of viral DNA and of viral structural proteins (Tooze, 1981).
The E1 region of adenovirus is the first region of adenovirus expressed after infection of the target cell This region consists of two transcriptional units, the E1A and E1B genes, which both are required for oncogenic transformation of primary (embryonal) rodent cultures. The main functions of the E1A gene products are.
i) to induce quiescent cells to enter the ceil cycle and resume cellular DNA synthesis, and
ii) to transcriptionally activate the E1B gene and the other early regions (E2, E3, E4) Transfection of primary cells with the E1A gene alone can induce unlimited proliferation (immortalization), but does not result in complete transformation. However, expression cf E1A in most cases results in induction of programmed cell death (apoptosis), and only occasionally immortalization is obtained (Jochemsen et al., 1987) . Co-expression of the E1B gene is required to prevent induction of apoptosis and for complete morphological transformation to occur. In established immortal cell lines, high level expression of E1A can cause complete transformation in the absence of E1B (Roberts et al., 1985). The E1B encoded proteins assist E1A in redirecting the cellular functions to allow viral replication. The E1B 55 kD and E4 33kD proteins, which form a complex that is essentially localized in the nucleus, function in
inhibiting the synthesis of host proteins and in
facilitating the expression of viral genes. Their main influence is to establisn selective transport of viral mRNAs from the nucleus to the cytoplasm, concomittantly with the onset of the late phase of infeçtion The E1B 21 kD protein is important for correct temporal control of the productive infection cycle, thereby preventing premature death of the host cell before the virus life cycle has been completed Mutant viruses incapable of expressing the E1B 21 kD gene-product exhibit a shortened infection cycle that is accompanied by excessive
degradation of host cell chromosomal DNA (deg-phenotype) and in an enhanced cytopathic effect (cyt-phenotype) (Telling et al., 1994). The deg and cyt pnenotypes are suppressed when in addition the E1A gene is mutated, indicating that these pnenotypes are a function of E1A
(White et al., 1988). Furthermore, the E1B 21 kDa protein slows down the rate by wmch E1A switches on the other viral genes It is not yet known through which mechanisms) E1B 21 kD quenches these E1A dependent functions.
Vectors derived from human adenoviruses, in which at least the E1 region has oeen deleted and replaced by a gene of interest, have oeen used extensively for gene therapy experiments in tne pre-clinical and clinical phase.
As stated before all adenovirus vectors currently used in gene therapy have a deletion in the E1 region, where novel genetic information can be introduced. The E1 deletion renders the recomoinant virus replication defective (Stratford-Perricaudet and Pemcaudet, 1991). We have demonstrated that recombinant adenoviruses are able to efficiently transfer recombinant genes to the rat liver and airway epithelium of rhesus monkeys (Bout et al., 1994b, Bout et al., 1994a) In addition, we (Vincent et al., 1996a, Vincent et al., 1996b) and others (see e.g Haddada et al., 1993) have observed a very efficient m vivo adenovirus mediated gene transfer to a variety of tumor cells in vitro and to solid tumors in animals models (lung tumors, glioma) and human xenografts in
immunodeflclent mice (lung) in vivo (reviewed by Blaese et al., 1995).
In contrast to for instance retroviruses,
adenoviruses a) do not integrate into the host cell genome; b) are able to infect non-dividing cells and c) are able to efficiently transfer recombinant genes in vivo (Brody and Crystal, 1994). Those features make
adenoviruses attractive candidates for in vivo gene transfer of, for instance, suicide or cytokine genes into tumor cells.
However, a problem associated with current
recombinant adenovirus technology is the possibility of unwanted generation of replication competent adenovirus (RCA) during the production of recombinant adenovirus (Lochmuller et al., 1994, Imler et al., 1996) This is caused by homologous recombination between overlapping sequences from the recombinant vector and the adenovirus constructs present in the complementing cell line, such as the 293 cells (Granam et al., 1977). RCA in batches to be used in clinical trials is unwanted because RCA i) will replicate in an uncontrolled fashion; ii) can complement replication defective recombinant adenovirus, causing uncontrolled multiplication of the recombinant adenovirus and iii) batches containing RCA induce significant tissue damage and hence strong pathological side effects
(Lochmuller et al., 1994). Therefore, batches to be used in clinical trials should be proven free of RCA (Ostrove, 1994). In one aspect of the invention this problem in virus production is solved in that we have developed packaging cells that have no overlapping sequences with a new basic vector and thus are suited for safe large scale production of recombinant adenoviruses one of the
additional problems associated with the use of recombinant adenovirus vectors is the host-defence reaction against treatment with adenovirus.
Briefly, recombinant adenoviruses are deleted for the E1 region (see above) . The adenovirus E1 products trigger the transcription of the other early genes (E2, E3, E4 ), which consequently activate expression of the late virus genes. Therefore, it was generally thought that E1 deleted vectors would not express any other adenovirus genes.
However, recently it has been demonstrated that some cell types are able to express adenovirus genes in the absence of E1 sequences. This indicates, that some cell types possess the machinery to drive transcription of adenovirus genes In particular, it was demonstrated that such cells synthesize E2A and late adenovirus proteins.
In a gene therapy setting, this means that transfer of the therapeutic recombinant gene to somatic cells not only results in expression of the therapeutic protein but may also result in the synthesis of viral proteins Cells that express adenoviral proteins are recognized and killed by Cytotoxic T Lymphocytes, which thus a) eradicates the transduced cells and b) causes inflammations (Bout et al 1994a; Engelnardt et al., 1993 Simon et al., 1993). As this adverse reaction is hampering gene therapy, several solutions to this problem have been suggested, such as a) using lmmunosuppressive agents after treatment, b) retainment of the adenovirus E3 region in the recombinant vector (see patent application EP 95202213) and c) and using ts mutants of human adenovirus, which have a point mutation in the E2A region (patent WO/28938)
However, these strategies to circumvent the immune response have their limitations.
The use of ts mutant recombinant adenovirus
diminishes the immune response to some extent, but was less effective in preventing pathological responses in the lungs (Engeihardt et al., 1994a). The E2A protein may induce an immune response by itself and it plays a pivotal role in the switch to the synthesis of late adenovirus proteins. Therefore, it is attractive to rnaxe recombinant adenoviruses wnich are mutated m the E2 region, rendering it temperature sensitive (ts), as has been claimed in patent application WO/28938.
A major drawoack of this system is the fact that, altnough the E2 protein is unstable at the non-permissive temperature, the immunogenic protein is still being synthesized. In addition, it is to be expected that the unstable protein does activate late gene expression, albeit to a low extent. ts125 mutant recomomant
adenoviruses have been tested, and prolonged recombinant gene expression was reported (Yang et al., 1994b;
Engeihardt et al., 1994a; Engelhardt et al., 1994b; Yang et al., 1995) . However, pathology in the lungs of cotton rats was still high (Engelhardt et al., 1994a;, indicating that the use of ts mutants results in only a partial improvement in recombinant adenovirus technology. Others (Fang et al., 1996) did not observe prolonged gene expression in mice and dogs using ts125 recombinant adenovirus. An additional difficulty associated with the use of ts125 mutant adenoviruses is that a high frequency of reversion is ooserved. These revertants are either real revertants or the result of second site mutations (Kruijer et al., 1983, Nicolas et al., 1981). Both types of revertants have an E2A protein that functions at normal temperature and have therefore similar toxicity as the wild-type virus.
In another aspect of the present invention we therefore delete E2A coding sequences from tne recombinant adenovirus genome and transfect these E2A sequences into the (packaging) cell lines containing E1 sequences to complement recombinant adenovirus vectors. Major hurdles in this approach are a) that E2A should be expressed to very high levels and b) that E2A protein is very toxic to cells.
The current invention in yet another aspect therefore discloses use of the ts125 mutant E2A gene, which produces a protein that is not anle to bind DNA sequences at the non permissive temperature. High levels of this protein may be maintained in the cells (because it is not toxic at this temperature) until tne switch to the permissive temperature is made. This can be combined witn placing the mutant E2A gene under the direction of an mducible promoter, such as for instance tet, methallothionem, steroid mducible promoter retmoic acid β-receptor or otner mducible systems. However in yet another aspect of the invention, the use or an mducible promoter to control the moment of production of toxic wild-type E2A is disclosed.
Two salient additional advantages of E2A-deleted recombinant adenovirus are the increased capacity to harpor heterologous sequences and the permanent selection for cells that express tne mutant E2A. This second advantage relates to the high frequency of reversion of ts125 mutation when reversion occurs in a cell line narooring ts125 E2A, this wi ll be lethal to tne cell.
Therefore there is a permanent selection for those cells that express the ts125 mutant E2A protein. In addition, as we in one aspect of the invention generate E2A-deleted recombinant aoenovirus, we will not have the problem of reversion m our adenoviruses.
In yet another aspect of the invention as a further improvement the use of non-human cell lines as packaging cell lines is disclosed.
For GMP production of clinical batches of recombinant viruses it is desirable to use a cell line that has been used widely for production of other biotechnology
products. Most of the latter cell lines are from monkey origin, which have been used to produce e.g. vaccines. These cells can not be used directly for the production of recombinant human adenovirus, as human adenovirus can not or only to low levels replicate in cells of monkey origin. A block in the switch of early to late phase of adenovirus lytic cycle is underlying defective replication. However, host range mutations in the human adenovirus genome are described (hr400 - 404) which allow replication of human viruses in monkey cells. These mutations reside in the gene encoding E2A protein (Klessig and Grodzicker, 1979; Klessig et al., 1984; Rice and Klessig, 1985) (Klessig et al., 1984). Moreover, mutant viruses have been described that harbor both the hr and temperature-sensitive ts125 phenotype (Brough et al., 1985; Rice and Klessig, 1985).
We therefore generate packaging cell lines of monkey origin (e.g. VERO, CV1) that harbor:
a. E1 sequences, to allow replication of E1/E2 defective adenoviruses, and
b. E2A sequences, containing the hr mutation and the ts 125 mutation, named ts400 (Brough et al., 1985; Rice and Klessig, 1985) to prevent cell death by E2A overexpression, and/or
c. E2A sequences, just containing the hr mutation, under the control of an inducible promoter, and/or
d. E2A sequences, containing the hr mutation and the ts 125 mutation (ts400), under the control of an inducible promoter
Furthermore we disclose the construction of novel and improved combinations of (novel and improved) packaging cell lines and (novel and improved) recombinant adenovirus vectors. We provide:
1. a novel packaging cell line derived from diploid human embryonic retinoblasts (HER) that harbors nt. 80 -
5788 of the Ad5 genome. This cell line, named 911, deposited under no 95062101 at the ECACC, has many characteristics that make it superior to the commonly used 293 cells (Fallaux et al., 1996). 2. novel packaging cell lines that express just E1A genes and not E1B genes.
Established cell lines (and not human diploid cells of which 293 and 911 cells are derived) are able to express E1A to high levels without undergoing
apoptotic cell death, as occurs in numan diploid cells that express E1A in the absence of E1B.
Such cell lines are able to trans-complement E1B- defective recombinant adenoviruses, because viruses mutated for E1B 21 kD protein are apie to complete viral replication even faster than wild-type
adenoviruses (Telling et al., 1994). The constructs are described in detail below, and graphically represented in Figures 1-5. The constructs are transfected into the different established cell lines and are selected for high expression of E1A. This is done by operatively linking a selectable marker gene (e.g. NEO gene) directly to the E1B promoter. The E1B promoter is transcriptionally activated by the E1A gene product and therefore resistance to the selective agent (e.g. G418 in the case NEO is used as the selection marker) results m direct selection for desired expression of the E1A gene
3. Packaging constructs that are mutated or deleted for E1B 21 kD, but just express the 55 kD protein.
4. Packaging constructs to be used for generation of
complementing packaging cell lines from diploid cells (not exclusively of human origin) without the need of selection with marker genes. These cells are
immortalized by expression of E1A. However, in this particular case expression of E1B is essential to prevent apoptosis induced by E1A proteins
Selection of E1 expressing cells is achieved by selection for focus formation (immortalization), as described for 293 cells (Graham et al., 1977) and 911 cells (Fallaux et al.,1996), that are E1-transformed human embryonic kidney (HEK) cells and human embryonic retmoblasts (HER), respectively.
5. After transfection of HER cells with construct pIG. E1B (Fig. 4), seven independent cell lines cculd be established. These cell lines were designated PER.C1, PER.C3, PER.C4, PER.C5, PER.C6, PER.C8 ar.d PER.C9. PER denotes PGK-E1-Retmoblasts. These cell lines express E1A and E1B proteins, are stable (e.g. PER.C6 for more than 57 passages) and complement E1 defective
adenovirus vectors. Yields of recombinant adenovirus obtained on PER cells are a little higher than obtained on 293 cells. One of these cell lines
(PER.C6) has been deposited at tne ECACC under number 96022940.
6. New adenovirus vectors with extended E1 deletions
(deletion nt. 459 - 3510) . Those viral vectors lack sequences homologous to E1 sequences in said packaging cell lines. These adenoviral vectors contain pIX promoter sequences and the pIX gene, as pIX (from its natural promoter sequences) can only b expressed from the vector and not by packaging cells (l.atsui et al., 1986, Hoeben and Fallaux, pers. comm.; Imler et al., 1996).
7. E2A expressing packaging cell lines preferably based on either E1A expressing established cell lines or E1A - E1B expressing diploid cells (see unoer 2 - 4). E2A expression is either under the control of an inducible promoter or the E2A ts125 mutant is driven by either an mducible or a constitutive promoter.
8. Recombinant adenovirus vectors as described before (see 6) but carrying an additional deletion of E2A sequences.
9. Adenovirus packaging cells from monkey origin that are able to trans-complement E1-defective recombinant adenoviruses. They are preferably co-transfected with pIG.E1AE1B and pIG.NEO, and selected for NEO
resistance. Such cells expressing E1A and E1B are able to transcomplement E1 defective recombinant human adenoviruses, but will do so inefficiently because of a block of the synthesis of late adenovirus proteins in cells of monkey origin (Klessig and Grodzicker, 1979). To overcome this problem, we generate
recombinant adenoviruses that harbor a host-range mutation in the E2A gene, allowing human adenoviruses to replicate in monkey cells. Such viruses are generated as described in Figure 12,-except DNA from a hr-mutant is used for homologous recombination.
10. Adenovirus packaging cells from monkey origin as
described under 9, except that they will also be co-transfected with E2A sequences harboring the hr mutation. This allows replication of human
adenoviruses lacking E1 and E2A (see under 8). E2A in these cell lines is either under the control of an mducible promoter or the tsE2A mutant is used. In the latter case, the E2A gene will thus carry both the ts mutation and the hr mutation (derived from ts400). Replication competent human adenoviruses have been described that harbor both mutations (Brough et al., 1985; Rice and Klessig, 1985).
A further aspect of the invention provides otherwise improved adenovirus vectors, as well as novel strategies for generation and application of such vectors and a method for the mtracellular amplification of linear DNA fragments in mammalian cells.
The so-called "minimal" adenovirus vectors according to the present invention retain at least a portion of the viral genome that is required for encapsidation of tne genome into virus particles (the encapsidation signal), as well as at least one copy of at least a functional part or a derivative of the Inverted Terminal Repeat (ITR), that is DNA sequences derived from the termini of the linear adenovirus genome. The vectors according to the present invention will also contain a transgene linked to a promoter sequence to govern expression of the transgene. Packaging of the so-called minimal adenovirus vector can be achieved by co-infection with a helper virus or, alternatively, with a packaging deficient replicating helper system as described below.
Adenovirus-derived DNA fragments that can replicate in suitable cell lines and that may serve as a packaging deficient replicating helper system are generated as follows. These DNA fragments retain at least a portion of the transcribed region of the "late" transcription unit of the adenovirus genome and carry deletions in at least a portion of the E1 region and deletions in at least a portion of the encapsidation signal In addition, these DNA fragments contain at least one copy of an inverted terminal repeat (ITR). At one terminus of the transfected DNA molecule an ITR is located The other end may contain an ITR, or alternatively, a DNA sequence that is
complementary to a portion of the same strand of the DNA molecule other than the ITR If, in the latter case, the two complementary sequences anneal, the free 3'-hydroxyl group of the 3' terminal nucleotide of the hairpm- structure can serve as a primer for DNA synthesis by cellular and/or adenovirus-encoded DNA polymerases, resulting in conversion into a double-stranded form of at least a portion of the DNA molecule Further replication initiating at the ITR will result in a linear double- stranded DNA molecule, that is flanked by two ITR's, and is larger than the original transfected DNA molecule (see Fig. 13). This molecule can replicate itself in the transfected cell by virtue of the adenovirus proteins encoded by the DNA molecule and the adenoviral and cellular proteins encoded by genes in the host-cell genome This DNA molecule can not be encapsidated due to its large size (greater than 39000 base pairs) or due to the absence or a functional encapsidation signal. This DNA molecule is intended to serve as a helper for the production of defective adenovirus vectors in suitable cell lines.
The invention also comprises a method for the amplification of linear DNA fragments of variable size in suitable mammalian cells. These DNA fragments contain at least one copy of the ITR at one of the termini of the fragment. The other end may contain an ITR, or
alternatively, a DNA sequence that is complementary to a portion of the same strand of the DNA molecule other than the ITR. If, in the latter case, the two complementary sequences anneal, the free 3'-hydroxyl group of the
3' terminal nucleotide of the hairpin-structure can serve as a primer for DNA synthesis by cellular and/or
adenovirus-encoded DNA polymerases, resulting in
conversion of the displaced stand into a double stranded form of at least a portion of the DNA molecule. Further replication initiating at the ITR will result in a linear double-stranded DNA molecule, that is flanked by two ITR's, which is larger than the original transfected DNA molecule. A DNA molecule that contains ITR sequences at both ends can replicate itself in transfected cells by virtue of the presence of at least the adenovirus E2 proteins (viz. the DNA-bmding protein (DBP), the
adenovirus DNA polymerase (Ad-pol), and the preterminal protein (pTP). The required proteins may be expressed from adenovirus genes on the DNA molecule itself, from
adenovirus E2 genes integrated in the host-cell genome, or from a replicating helper fragment as described above.
Several groups have shown that the presence of ITR sequences at the end of DNA molecules are sufficient to generate adenovirus mmichromosomes that can replicate, if the adenovirus-proteins required for replication are provided in trans e.g. by infection with a heipervirus (Hu et al., 1992); (Wang and Pearson, 1985) ; (Hay et al., 1984) Hu et al . , (1992) observed the presence and replication of symmetrical adenovirus minichromosome- dimers after transfection of plasmids containing a single ITR. The authors were able to demonstrate that these dimeric mmichromosomes arize after tail-to-tail ligation of the single ITR DNA molecules In DNA extracted from defective adenovirus type 2 particles, dimeric molecules of various sizes have also been observed using electron- microscopy (Daniell, 1976). It was suggested that the incomplete genomes were formed by illegitimate
recombination between different molecules and that variations in the position of the sequence at which the illegitimate base pairing occurred were resonsible for the heterogeneous nature of the incomplete genomes. Based on this mechanism it was speculated that, in theory,
defective molecules with a total length of up to two times the normal genome could be generated. Such molecules could contain duplicated sequences from either enc of the genome. However, no DNA molecules larger than the full- length virus were found packaged in the defective
particles (Daniell, 1976). This can be explained by the size-limitations that apply to the packaging In addition, it was observed that in the virus particles DNA-molecules with a duplicated left-end predominated over those
containing the right-end terminus (Daniell, 1976). This is fully explained by the presence of the encapsidation signal near that left-end of the genome (Grable and
Hearing, 1990; Grable and Hearing, 1992; Hearing et al., 1987).
The major problems associated with the current adenovirus-derived vectors are:
A) The strong lmmunogenicity of the virus particle B) The expression of adenovirus genes that reside in the adenoviral vectors, resulting in a Cytotoxic T-cell response against the transduced cells.
C) The low amount of heterologous sequences that can oe accommodated in the current vectors (Up to
maximally approx. 8000 bp. of heterologous DNA). Ad A) The strong immunogenicity of the adenovirus particle results m an immunological response of the host, even after a single administration of the adenoviral vector. As a result of the development of neutralizing antibodies, a subsequent administration of the virus will be less effective or even completely ineffective. However, a prolonged or persistent expression of the transferred genes will reduce the number of administrations required and may bypass the problem.
Ad B) Experiments performed by Wilson and
collaborators have demonstrated that after adenovirus - mediated gene transfer into immunocompetent animals, the expression of the transgene gradually decreases and disappears approximately 2 - 4 weeks post- infection 'Yang et al., 1994a, Yang et al., 1994b). This is caused by the development of a Cytotoxic T-Cell (CTL) response against the transduced cells. The CTLs were directed against adenovirus proteins expressed by the viral vectors. In the transduced cells synthesis of the adenovirus DNA-bmding protem (the E2A-gene product), penton and fiber proteins (late-gene products) could be established. These
adenovirus proteins, encoded by the viral vector, were expressed despite deletion of the E1 region. This
demonstrates that deletion of the E1 region is not sufficient to completely prevent expression of the viral genes (Engelhardt et al., 1994a).
Ad C) Studies by Graham and collaborators have demonstrated that adenoviruses are capable of
encapsidatmg DNA of up to 105% of the normal genome size (Bett et al., 1993). Larger genomes tend to be mstable resulting in loss of DNA sequences during propagation of the virus. Combining deletions m the E1 and E3 regions of tne virual genomes increases the maximmum size of the foreign that can be encapsidated to approx. 8.3 kb. In addition, some sequences of the E4 region appear to be dispensable for virus growth (adding another 1.8 kb to the maximum encapsidation capacity). Also the E2A region can be deleted from the vector, when the E2A gene product is provided in trans in the encapsidation cell line, adding another 1.6 kb. It is, however, unlikely that the maximum capacity of foreign DNA can be significantly increased further than 12 kb.
We developed a new strategy for the generation and production of helperfree-stocks of recombinant adenovirus vectors that can accomodate up to 38 kb of foreign DNA. Only two functional ITR sequences, and sequences that can function as an encapsidation signal need to be part of the vector genome. Such vectors are called minimal
adenovectors. The helper functions for the minimal adenovectors are provided in trans by encapsidation defective-replication competent DNA molecules that contain all the viral genes encoding the required gene products, with the exception of those genes that are present in the host- cell genome, or genes that reside in the vector genome.
The applications of the disclosed inventions are outlined below and will be illustrated in the experimental part, which is only intended for said purpose, and should not be used to reduce the scope of the present invention as understood by the person skilled in the art. Use of the IG packaging constructs Diploid cells.
The constructs, in particular pIG.E1A.E1B, will be used to transfect diploid human cells, such as Human Embryonic Retinoblasts (HER), Human Embryonic Kidney cells (HEK), and Human Embryonic Lung cells (HEL). Transfected cells will be selected for transformed phenotype (focus formation) and tested for their ability to support propagation of E1-deleted recombinant adenovirus, such as IG.Ad.MLPI.TK. Such cell lines will be used for the generation and (large-scale) production of E1-deleted recombinant adenoviruses. Such cells, infected with recombinant adenovirus are also intended to be used in vivo as a local producer of recombinant adenovirus, e.g. for the treatment of solid tumors.
911 cells are used for the titration, generation and production of recombinant adenovirus vectors (Fallaux et al., 1996).
HER cells transfected with pIG.E1A.E1B has resulted in 7 independent clones (called PER cells). These clones are used for the production of E1 deleted (including non- overlapping adenovirus vectors) or E1 defective
recombinant adenovirus vectors and provide the basis for introduction of e.g. E2B or E2A constructs (e.g. ts125E2A, see below), E4 etc., that will allow propagation of adenovirus vectors that have mutations in e.g. E2A or E4.
In addition, diploid cells of otner species that are permissive for human adenovirus, such as the cotton rat
(Sigmodon nispidus) (Pacini et al., 1984), Syrian hamster (Morin et al., 1987) or chimpanzee (Levrero et al., 1991), will be immortalized with these constructs. Such cells, infected with recombinant adenovirus, are also intended to be used in vivo for the local production of recombinant adenovirus, e.g. for the treatment of solid tumors.
Established cells. The constructs, in particular pIG.E1A.NEO, can be used to transfect established cells, e.g. A549 (human bronchial carcinoma), KB (oral carcinoma), MRC-5 (human diploid lung cell line) or GLC cell lines (small cell lung cancer) (de Leij et al., 1985; Postmus et al., 1988) and selected for NEO resistance. Individual colonies of resistant cells are isolated and tested for their capacity to support propagation of E1 -deleted recombinant
adenovirus, such as IG.Ad.MLPI.TK. When propagation of E1 deleted viruses on E1A containing cells is possible, such cells can be used for the generation and production of E1-deleted recombinant adenovirus. They are also be used for the propagation of E1A deleted/E1B retained
recombinant adenovirus.
Established cells can also be co-transfected with pIG.E1A.E1B and pIG.NEO (or another NEO containing expression vector) Clones resistant to G418 are tested for their ability to support propagation of II deleted recombinant adenovirus, such as IG.Ad.MLPI.TK and used for the generation and production of E1 deleted recombinant adenovirus and will be applied m vivo for lccal
production of recombinant virus, as describee for the diploid cells (see above).
All cell lines, including transformed diploid cell lines or NEO-resistant established lines, can be used as the basis for the generation of 'next generation'
packaging cells lines, that support propagation of
E1-defective recombinant adenoviruses that also carry deletions in other genes, such as E2A and E4. Moreover, they will provide the basis for the generation of minimal adenovirus vectors as disclosed herein.
E2 expressing cell lines
Packaging cells expressing E2A sequences are and will be used for the generation and (large scale) production of E2A-deieted recombinant adenovirus.
The newly generated human aoenovirus packaging cell lines or cell lines derived from species permissive for human adenovirus (E2A or ts125E2A; E1A + E2A; E1A + E1B + E2A; E1A + E2A/ts125, E1A + E1B + E2A/ts125) or non- permissive cell lines such as monkey cells (hrE2A or hr + ts125E2A; E1A + hrE2A; E1A + E1B + hrE2A; E1A +
hrE2A/ts125; E1A + E1B + hrE2A/ts125) are and will be used for the generation and (large scale) production of E2A deleted recombinant adenovirus vectors In addition, they will be applied in vivo for local production of
recombinant virus, as described for the diploid cells (see above) . Novel adenovirus vectors.
The newly developed adenovirus vectors narboring an E1 deletion of nt. 459-3510 will be used for gene transfer purposes. These vectors are also the basis for the development of further deleted adenovirus vectors that are mutated for e.g. E2A, E2B or E4. Such vectors will be generated e.g. on the newly developed packaging cell lines described above (see 1-3).
Minimal adenovirus packaging system
We disclose adenovirus packaging constructs (to be used for the packaging of minimal adenovirus vectors may nave the following characteristics:
a. the packaging construct replicates
b. the packaging construct can not be packaged because the packaging signal is deleted
c. the packaging construct contains an internal hairpm- forming sequence (see section 'Experimental, suggested hairpin' see Fig. 15)
d. because of the internal hairpin structure, the
packaging construct is duplicated, that is the DNA of the packaging construct becomes twice as long as it was before transfection into the packaging cell (in our sample it duplicates from 35 kb to 70 kb ) . This duplication also prevents packaging. Note that this duplicated DNA molecule has ITR's at both termini (see e.g. Fig. 13)
e. this duplicated packaging molecule is able to
replicate like a 'normal adenovirus' DNA molecule f. the duplication of the genome is a prerequisite for the production of sufficient levels of adenovirus proteins, required to package the minimal adenovirus vector g. the packaging construct has no overlapping sequences with the minimal vector or cellular sequences that may lead to generation of RCA by homologous
recombination.
This packaging system will be used to produce minimal adenovirus vectors. The advantages of minimal adenovirus vectors e.g. for gene therapy of vaccination purposes, are well known (accommodation of up to 38 kb; gutting of all potentially toxic and lmmunogenic adenovirus genes).
Adenovirus vectors containing mutations in essential genes (including minimal adenovirus vectors) can also be propagated using this system. Use of intracellular E2 expressing vectors.
Minimal adenovirus vectors are generated using the helper functions provided in trans by packaging-deficient replicating helper molecules. The adenovirus-derived ITR sequences serve as origins of DNA replication in the presence of at least the E2-gene products. When the E2 gene products are expressed from genes in the vector genome (N.B the gene (s) must be driven by an E1- independent promoter), the vector genome can replicate in the target cells. This will allow an significantly increased number of template molecules in the target cells, and, as a result an increased expression of the genes of interest encoded by the vector. This is of particular interest for approaches of gene therapy in cancer.
Applications of intracellular amplification of linear DNA fragments. A similar approach could also be taken if
amplification of linear DNA fragments is desired DNA fragments of known or unknown sequence could be amplified in cells containing the E2-gene products if at least one ITR sequence is located near or at its terminus. There are no apparent constraints on the size of the rragment. Even fragments much larger than the adenovirus genome (36 kb) should be amplified using this approach. It is thus possible to clone large fragments in mammalian cells without either shuttling the fragment into bacteria (such as E. coli) or use the polymerase chain reaction (P.C.R.). At the end stage of an productive adenovirus infection a single cell can contain over 100,000 copies of the viral genome. In the optimal situation, the linear DNA fragments can be amplifιed to similar levels Thus, one should be able to extract more than 5 μg of DNA fragment per 10 million cells (for a 35-kbp fragment). This system can be used to express heterologous proteins (equivalent to the
Simian Virus 40-based COS-cell system) ror research or for therapeutic purposes In addition, the system can be used to identify genes in large fragments of DNA. Random DNA fragments may be amplified (after addition of ITRs) and expressed during mtracellular amplification. Election or, selection of those cells with the oesired pnenotvpe can be used to enrich the fragment of interest and to isolate the gene. EXPERIMENTAL
Generation of cell lines able to transcomplement E1 defective recombinant adenovirus vectors.
1. 911 cell line
We have generated a cell line that harbors E1 sequences of adenovirus type 5, able to trans-complement E1 deleted recombinant adenovirus (Fallaux et al., 1996).
This cell line was obtained by transrection of human diploid human embryonic retinoblasts (HER) with pAd5XhoIC, that contains nt. 80 - 5788 of Ad5; one of the resulting transformants was designated 911. This cell line has been shown to be very useful in the propagation of E1 defective recombinant adenovirus. It was found to be superior to the 293 cells. Unlike 293 cells, 911 cells lack a fully transformed pnenotvpe, which most likely is the cause of performing better as adenovirus packaging line:
plaque assays can be performed faster (4 - 5 days instead of 8-14 days on 293)
monolayers of 911 cells survive better under agar overlay as required for plaque assays
higher amplification of E1 -deleted vectors
In addition, unlike 293 cells that were transfected with sheared adenoviral DNA, 911 cells were transfected using a defined construct. Transfection efficiencies of 911 cells are comparable to those of 293.
New packaging constructs.
Source of adenovirus sequences.
Adenovirus sequences are derived either from
pAd5. Sa1B, containing nt. 80 - 9460 of human adenovirus type 5 (Bernards et al., 1983) or from wild-type Ad5 DNA. pAd5. Sa1B was digested with Sall and Xhol and the large fragment was religated and this new clone was named pAd5. X/S.
The pTN construct (constructed by Dr. R. Vogels,
IntrcGene, The Netherlands) was used as a source for the human PGK promoter and the NEO gene.
Human PGK promoter and NEOR gene.
Transcription of E1A sequences in the new packaging constructs is driven by the human PGK promoter (Michelson et al., 1983; Singer-Sam et al., 1984), derived from plasmid pTN (gift of R. Vogels), which uses pUC119 (Vieira and Messing, 1987) as a backbone. This plasmid was also used as a source for NEO gene fused to the Hepatitis B Virus (HBV) poly-adenylation signal. Fusion of PGK promoter to E1 genes (Fig. 1)
In order to replace the E1 sequences of Ad5 (ITR, origin of replication and packaging signal) by
heterologous sequences we have amplified E1 sequences
(nt.459 to nt. 960) of Ad5 by PCR, using primers Eal and Ea2 (see Table I). The resulting PCR product was digested with Clal and ligated into Bluescript (Stratagene), predigested with Clal and EcoRV, resulting in construct pBS.PCRI.
Vector pTN was digested with restriction enzymes EcoRI (partially) and Seal, and the DNA fragment
containing the PGK promoter sequences was ligated into PBS PCRI digested with Seal and EcoRI. The resulting construct PBS.PGK.PCRI contains the human PGK promoter operatively linked to Ad5 E1 sequences from nt 459 to nt. 916.
Construction of pIG.E1A.E1B.X (Fig. 2) pIG.E1A.E1B.X was made by replacing the Scal-BspEI fragment of pAT-X/S by the corresponding fragment from PBS.PGK.PCRI (containing the PGK promoter linked to E1A sequences).
pIG.E1A.E1B.X contains the E1A and E1B coding sequences under the direction of the PGK promoter.
As Ad5 sequences from nt.459 to nt 5788 are present in this construct, also pIX protein of adenovirus is encoded by this plasmid.
Construction of pIG.E1A.NEO (Fig. 3)
In order to introduce the complete E1B promoter and to fuse this promoter in such a way that the AUG codon of E1B 21 kD exactly functions as the AUG codon of NEOR, we amplified the E1B promoter using primers Ea3 and Ep2, where primer Ep2 introduces an Ncol site in the PCR fragment. The resulting PCR fragment, named PCRII, was digested with Hpal and Ncol and ligated into pAT-X/S, which was predigested with Hpal and with Ncol The resulting plasmid was designated pAT-X/S-PCR2. The Ncol - StuI fragment of pTN, containing the NEO gene and part of the Hepatitis B Virus (HBV) poly-adenylation signal, was cloned into pAT-X/S-PCR2 (digested with Ncol and Nrul). The resulting construct: pAT-PCR2-NEO. The poly- adenylation signal was completed by replacing the Scal- Sall fragment of pAT-PCR2-NEO by the corresponding fragment of pTN (resulting in pAT.PCR2.NEO.p(A)). The Seal - Xbal of pAT.PCR2.NEO.p (A) was replaced by the
corresponding fragment of pIG.E1A.E1B-X, containing the PGK promoter linked to E1A genes.
The resulting construct was named pIG.E1A.NEO, and thus contains Ad5 E1 sequences (nt 459 to nt 1713) under the control of the human PGK promoter.
Construction of pIG.EIA.E1B (Fig. 4) pIG.E1A.E1B was made by amplifying the sequences encoding the N-terminal ammo acids of E1B 55kd using primers Eb1 and Eb2 (introduces a Xhol site). The
resulting PCR fragment was digested with Bglll and cloned into Bglll/Nrul of pAT-X/S, thereby obtaining pAT-PCR3. pIG.E1A.E1B was constructed by introducing the HBV poly(A) sequences of pIG.E1A.NEO downstream of E1B sequences of pAT-PCR3 by exchange of Xbal - Sall fragment of pig.E1A.NEO and the Xbal Xhol fragment of pAT PCR3
pIG.EIA.E1B contains nt. 459 to nt. 3510 of Ad5, that encode the E1A and E1B proteins. The E1B sequences are terminated at the splice acceptor at nt.3511. No pIX sequences are present in this construct. Construction of pIG.NEO (Fig. 5) pIG NEO was generated by cloning the Hpal - Seal fragment of pIG.E1A.NEO, containing the NEO gene under the control of the Ad.5 E1B promoter, into pBS digested with EcoRV and Seal.
This construct is of use when established cells are transfected with E1A. E1B constructs and NEO selection is required. Because NEO expression is directed by the E1B promoter, NEO resistant cells are expected to co-express E1A, which also is advantageous for maintaining high levels of expression of E1A during long-term culture of the cells. Testing of constructs.
The integrity of the constructs pIG.E1A.NEO,
pIG.E1A.E1B.X and pIG.E1A.E1B was assessed by restriction enzyme mapping, furthermore, parts of the constructs that were obtained by PCR analysis were confirmed by sequence analysis No changes in the nucleotide sequence were found.
The constructs were transfected into primary BRK (Baby Rat Kidney) cells and tested for their ability to immortalize (pIG.E1A.NEO) or fully transform
(pAd5.XhoIC, pIG.E1A.E1B X and pIG.E1A.E1B) these cells.
Kidneys of 6-day old WAG-Rij rats were isolated, homogenized and trvpsmized. Subconfluent disnes (diameter 5 cm) of the BRK cell cultures were transfected with 1 or 5 μg of pIG.NEO, pIG.E1A.NEO, pIG.E1A.E1B, pIG.E1A.E1B.X, pAd5XhoIC, or with pIG.E1A.NEO together with PDC26
(Van der E1sen et al., 1983), carrying the Ad5.E1B gene under control of the SV40 early promoter. Three weeks post-transfection, when foci were visible, the dishes were fixed, Giemsa stained and the foci counted.
An overview of the generated adenovirus packaging constructs, and their ability to transform BRK, is presented in Fig. 6. The results indicate that the constructs pIG.E1A.E1B and pIG.E1A.E1B.X are able to transform BRK cells in a dose-dependent manner. The efficiency of transformation is similar fcr both
constructs and is comparable to what was found with the construct that was used to make 911 cells, namely
pAd5.XhoIC.
As expected, pIG.E1A.NEO was hardly able to
immortalize BRK. However, co-transfection of an E1B expression construct (PDC26) did result m a significant increase of the number of transformants (18 versus 1), indicating that E1A encoded by pIG.E1A.NEO is functional.
We conclude therefore, that tne newly generated packaging constructs are suited fcr the generation of new adenovirus packaging lines.
Generation of cell lines with new packaging
constructs Cell lines and cell culture Human A549 bronchial carcinoma cells (Shapiro et al., 1978), human embryonic retinoblasts (HER), Ad5-E1- transfor-med human embryonic kidney (HEK) cells (293;
Graham et al., 1977) cells and Ad5-transformed HER cells (911; Fallaux et al, 1996)) and PER cells were grown in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% Fetal Calf Serum (FCS) and antibiotics in a 5% CO2 atmospnere at 37°C. Cell culture media, reagents and sera were purchased from Gibeo Laboratories (Grand Island, NY). Culture plastics were purchased from Gremer (Nurtmgen, Germany) and Corning (Corning, NY).
Viruses and virus techniques
The construction of adenoviral vectors
IG.Ad.MLP.nls.lacZ, IG.Ad.MLP.luc, IG.Ad.MLP.TK and
IG.Ad.CMV.TK is described in detail in patent application
EP 95202213. The recombinant adenoviral vector IG.Ad.MLP.nls.lacZ contains the E.coli lacZ gene, encoding β-gaiactosidase, under control of the Ad2 major late promoter
(MLP).IG.Ad.MLP.luc contains the firefly luciferase gene driven by the Ad2 MLP Adenoviral vectors IG.Ad.MLP.TK and IG.Ad.CMV.TK contain the Herpes Simplex Virus thymidine kmase (TK) gene under the control of the Ad2 MLP and the Cytomegalovirus (CMV) enhancer/promoter, respectively. Transfections
All transfections were performed by caicium-pnosphate precipitation DNA (Graham and Van der Eb, 1973) with the GIBCO Calcium Phosphate Transfection System (GIBCO BRL Life Technologies Inc., Gaithersburg, USA), according to the manufacturers protocol.
Western blotting Subconfluent cultures of exponentially growing
293,911 and Ad5-E1-transformed A549 and PER cells were cashed with PBS and scraped in Fos-RIPA buffer (10 mM Tris (pH 7,5), 150 mM NaCl, 1% NP40,01% sodium dodecyl sulphate (SDS), 1% NA-DOC, 0,5 mM phenyl methyl sulphonyl fluoride PMSF), 0,5 mM trypsin inhibitor, 50 mM NaF and 1 mM sodium vanadate). After 10 min. at room temperature, lysates were cleared by centrifugation. Protein
concentrations were measured with the Biorad protein assay kit, and 25 μg total cellular protein was loaded on a 12 5% SDS-PAA gel. After electrophoresis, proteins were transferred to nitrocellulose (lh at 300 mA). Prestamed standards (Sigma, USA) were run in parallel. Filters were blocked with 1% bovine serum albumin (BSA) in TBST (10 mM Tris, pH 8, 15 mM NaCl, and 0.05% Tween-20) for 1 hour. First antibodies were the mouse monoclonal anti-Ad5-E1B- 55-kDA antibody A1C6 (Zantema et al., unpublished), the rat monoclonal anti-Ad5-E1B-221-kDa antibody ClGll (Zantema et al., 1985). The second antibody was a
horseradish peroxidase-labeled goat anti-mouse antibody (Promega). Signals were visualized by enhanced
chemolummescence (Amersham Corp, UK).
Southern blot analysis
High molecular weight DNA was isolated and 10 μg was digested to completion and fractionated on a 0.7% agarose gel. Southern blot transfer to Hybond N+ (Amersham, UK) was performed with a 0.4 M NAOH, 0.6 M NaCl transfer solution (Church and Gilbert, 1984). Hybridization was performed with a 2463-nt Sspl-Hmdlll fragment from pAd5. SalB (Bernards et al., 1983). This fragment consists of Ad5 bp. 342-2805. The fragment was radiolabeled with α-32P-dCTP with the use of random hexanucleotide primers and Klenow DNA polymerase. The southern blots were exposed to a Kodak XAR-5 film at -80°C and to a Phospho-Imager screen which was analyzed by B&L systems Molecular
Dynamics software.
A549
Ad5-E1-transformed A549 human bronchial carcinoma cell lines were generated by transfection with pIG.E1A.NEO and selection for G418 resistance. Thirty-one G418 resistant clones were established. Co-transfection of pIG.EIA.E1B with pIG.NEO yielded seven G418 resistant cell lines.
PER
Ad5-E1-transformed human embryonic retina (HER) cells were generated by transfection of primery HER cells with plasmid pIG.EIA.E1B. Transformed cell lines were
established from well-separated foci. We were able to establish seven clonal cell lines, which we called PER.C1, PER.C3, PER.C4, PER.C5, PER.C6, PER.C8 and PER.C9.
One of the PER clones, namely PER.C6, has been deposited at the ECACC under number 96022940.
Expression of Ad5 E1A and E1B genes in transformed A549 and PER cells
Expression of the Ad5 E1A and the 55-kDa and 21 kDa E1B proteins in the established A549 and PER cells was studied by means of Western blotting, with the use of monoclonal antibodies (mAb). Mab M73 recognizes the E1A products, whereas Mabls AIC6 and ClGll are directed against the 55 -kDa and 21 kDa E1B proteins, respectively.
The antibodies did not recognize proteins in extracts from the parental A549 or the primary HER cells (data not shown). None of the A549 clones that were generated by co-transfection of pIG.NEO and pIG.E1A.E1B expressed detectable levels of E1A or E1B proteins (not shown). Some of the A549 clones that were generated by transfection with pIG.E1A.NEO expressed the Ad5 E1A proteins (Fig. 7), but the levels were much lower than those detected in protein lysates from 293 cells. The steady state E1A levels detected in protein extracts from PER cells were much higher than those detected m extracts from A549- derived cells All PER cell lines expressed similar levels of E1A proteins (Fig. 7). The expression of the E1B proteins particularly in the case of E1B 55 kDa, was mote variable. Compared to 911 and 293, the majority of the PER clones express high levels of E1B 55 kDa and 21 kDa. The steady state level of E1B 21 kDa was the highest in
PER.C3. None of the PER clones lost expression of the Ad5 E1 genes upon serial passage of the cells (not shown). We found that the level of E1 expression m PER cells remained stable for at least 100 population doublings.
We decided to characterize the PER clones in more detail. Southern analysis of PER clones
To study the arrangement of the Ad5-E1 encoding sequences in the PER clones we performed Southern
analyses. Cellular DNA was extracted from all PER clones, and from 293 and 911 cells. The DNA was digested with Hindlll, which cuts once m the Ad5 E1 region. Southern hybridization on Hindlll-digested DNA, using a
radiolaoeled Ad5-E1-specific probe revealed the presence of several integrated copies of pIG.EIA.E1B in the genome of the PER clones. Figure 8 shows the distribution pattern of E1 sequences in the high molecular weignt DNA of the different PER cell lines. The copies are concentrated in a single band, which suggests that they are integrated as tandem repeats. In the case of PER. C3, C5, C6 and C9 we found additional hybridizing bands of low molecular weight that indicate the presence of truncated copies of
pIG.E1A.E1B. The number of copies was determined with the use of a Phospho-Imager. We estimated that PER. C1, C3, C4, C5, C6, C8 and C9 contain 2, 88, 5,4, 5, 5 and 3 copies of the Ad5 E1 coding region, respectively, and that 911 and 293 cells contain 1 and 4 copies of the Ad5 E1 sequences, respectively. Transfection efficiency
Recombinant adenovectors are generated by cotransfection of adaptor plasmids and the large Clal fragment of Ad5 into 293 cells (gee patent application EP 95202213). The recombinant virus DNA is formed by homologous recombination between the homologous viral sequences that are present in the plasmid and the
adenovirus DNA. The efficacy of this method, as well as that of alternative strategies, is highly dependent on the transfectability of the helper cells. Therefore, we compared the transfection efficiencies of some of the PER clones with 911 cells, using the E.coli
β-galactosidase-encoding lacZ gene as a reporter (Fig. 9).
Production of recombinant adenovirus
Yields of recombinant adenovirus obtained after inoculation of 293, 911, PER.C3, PER.C5 and PER.C6 with different adenovirus vectors are presented m Table II.
The results indicate that the yields obtained on PER cells are at least as high as those obtained on the existing cell lines.
In addition, the yields of the novel adenovirus vector IG.Ad.MLPI.TK are similar or higher than the yields obtained for the other viral vectors on all cell lines tested.
Generation of new adenovirus vectors (Fig. 10).
The used recombinant adenovirus vectors (see patent application on EP 95202213) are deleted for E1 sequences from 459 to nt. 3328.
As construct pE1A. E1B contains Ad5 sequences 459 to nt. 3510 there is a sequence overlap of 183 nt. between E1B sequences in the packaging construct pIG.E1A.E1B and recombinant adenoviruses, sucn as e.g. IG.Ad.MLP.TK. The overlapping sequences were deleted from the new adenovirus vectors. In addition, non-coding sequences derived from lacZ, that are present in the original contructs, were deleted as well. This was achieved (see Fig. 10) by PCR amplification of the SV40 poly (A) sequences from pMLP.TK using primers SV40-1 (introduces a BamHI site) and SV40-2 (introduces a Bglll site). In addition, Ad5 sequences present in this construct were amplified from nt 2496 (Ad5, introduces a Bglll site) to nt. 2779 (Ad5-2). Both PCR fragments were digested with Bglll and were ligated. The ligation product was PCR amplified using primers SV40-1 and Ad5-2. The PCR product obtained was cut with BamHI and Aflll and was ligated into pMLP.TK predigested with the same enzymes. The resulting construct, named pMLPI.TK, contains a deletion in adenovirus E1 sequences from nt 459 to nt 3510.
Packaging system
The combination of the new packaging construct pIG.EIA.E1B and the recombinant adenovirus pMLPI.TK, which do not have any sequence overlap, are presented m Fig. 11. In this figure, also the original situation is presented, where the sequence overlap is indicated.
The absence or overlapping sequences between
pIG.E1A.E1B and pMLPI.TK (Fig. 11a) excludes the
possibility of homologous recombination between packaging construct and recombinant virus, and is therefore a significant improvement for production of recombinant adenovirus as compared to the original situation.
In Fig. 11b the situation is depicted for pIG.E1A.NEO and IG.Ad.MLPI TK. pIG.E1A.NEO when transfected into established cells, is expected to be sufficient to support propagation of E1-deleted recombinant adenovirus. This combination does not have any sequence overlap, preventing generation of RCA by homologous recombination. In
addition, this convenient packaging system allows the propagation of recombinant adenoviruses that are deleted just for E1A sequences and not for E1B sequences.
Recombinant adenoviruses expressing E1B m the absence of E1A are attractive, as the E1B protein, in particular E1B 19kD, is able to prevent infected human cells from lysis by Tumor Necrosis Factor (TNF) (Goodmg et al., 1991) .
Generation of recombinant adenovirus derived from pMLPI.TK.
Recombinant adenovirus was generated by cotransfection of 293 cells with Sail linearized pMLPI.TK DNA and Clal linearized Ad5 wt DNA. The procedure is schematically represented in Fig. 12.
Outline of the strategy to generate packaging systems for minimal adenovirus vector
Name convention of the plasmids used: p plasmid
I ITR (Adenovirus Inverted Terminal Repeat)
C Cytomegalovirus (CMV) Enhancer/Promoter Combination L Firefly Luciferase Coding Sequence hachaw Potential hairpin that can be formed after digestion with
restriction endonuclease Asp718 in its correct and in the. reverse orientation, respectively (Fig. 15).
Eg. pICLhaw is a plasmid that contains the adenovirus ITR followed by the CMV-driven luciferase gene and the Asp718 hairpin m the reverse (non-functional)
orientation.
1.1 Demonstration of the competence of a synthetic DNA sequence, that is capable of forming a hairpin- structure, to serve as a primer for reverse strand
synthesis for the generation of double-stranded DNA molecules in cells that contain and express adenovirus genes.
Plasmids pICLhac, pICLhaw, pICLI and pICL were generated using standard techniques. The schematic representation of these plasmids is shown in Figs. 16-19.
Plasmid pICL is derived from the following plasmids: nt.1 - 457 pMLP10 (Levrero et al., 1991)
nt.458 - 1218 pCMVβ (Clontech, EMBL Bank No. U02451) nt.1219 - 3016 pMLP . luc (IntroGene, unpublished) nt.3017 - 5620 pBLCAT5 (Stem and Whelan, 1989) The plasmid has been constructed as follows:
The tet gene of plasmid pMLP10 has been inactivated by deletion of the BamHI-Sail fragment, to generate pMLP10ΔSB. Using primer set PCR/MLP1 and PCR/MLP3 a 210 bp fragment containing the Ad5-ITR, flanked by a synthetic Sall restriction site was amplified using pMLP10 DNA as the template. The PCR product was digested with the enzymes EcoRI and SgrAI to generate a 196 bp. fragment. Plasmid pMLP10ΔSB was digested with EcoRI and SgrAI to remove the ITR. This fragment was replaced by the EcoRI- SgrAI-treated PCR fragment to generate pMLP/SAL .
Plasmid pCMV-Luc was digested with PvuII to completion and recirculated to remove the SV40-derived poly-adenylation signal and Ad5 sequences with exception of the Ad5
left-terminus. In the resulting plasmid, pCMV-lucΔAd, the
Ad5 ITR was replaced by the Sal-site-flanked ITR from plasmid pMLP/SAL by exchanging the Xmnl-SacII fragments. The resulting plasmid, pCMV-lucΔAd/SAL, the Ad5 left terminus and the CMV-driven luciferase gene were isolated as an Sall-Smal fragment and inserted in the Sall and Hpal digested plasmid pBLCATS, to form plasmid pICL. Plasmid pICL is represented in Fig 19; its sequence is presented in Fig. 20.
Plasmid pICL contains the following features nt. 1-457 Ad5 left terminus (Sequence 1-457 of
human adenovirus type 5)
nt. 458-969 Human cytomegalovirus enhancer and
immediate
early promoter (Boshart et al., 1985) (from plasmid pCMVβ,
Clontech, Palo Alto, USA) nt. 970-1204 SV40 19S exon and truncated 16/19S intron
(from plasmid pCMVβ)
nt. 1218-2987 Firefly luciferase gene (from pMLP.luc) nt. 3018-3131 SV40 tandem poly-adenylation signals from late transcript, derived from piasmid pBLCAT5)
nt. 3132-5620 pUC12 backbone (derived from plasmid
pBLCAT5 )
nt. 4337-5191 β-lactamase gene (Amp-resistence gene, reverse orientation)
Plasmid pICLhac and pICLhaw
Plasmids pICLhac and pICLhaw were derived from plasmid pICL by digestion of the latter plasmid with the restriction enzyme Asp718 The linearized plasmid was treated with Calf-Intestine Alkaline Phosphatase to remove the 51 phoshate groups. The partially complementary synthetic single-stranded oligonucleotide Hp/aspi en Hp/asp2 were annealed and phosphorylated on their 5' ends using T4-polynucleotιde kmase.
The phosporylated double-stranded oligomers were mixed with the dephosporylated pICL fragment and ligated. Clones containing a single copy of the synthetic oligonucleotide inserted into the plasmid were isolated and characterized using restriction enzyme digests. Insertion of the oligonucleotide into the Asp718 site will at one junction recreate an Asp718 recognition site, whereas at the other junction the recognitionsite will be disrupted. The orientation and the integrity of the inserted
oligonucleotide was verified in selected clones by sequence analyses. A clone containing the oligonucleotide in the correct orientation (the Asp718 site close to the 3205 EcoRI site) was denoted pICLhac. A clone with the oligonucleotide in the reverse orientation (the Asp718 site close to the SV40 derived poly signal) was designated pICLhaw. Plasmids pICLhac and pICLhaw are represented in Figs 16 and 17.
Plasmid pICLI was created from plasmid pICL by insertion of the Sall-SgrAI fragment rrom pICL, containing the Ad5-ITR into the Asp718 site of pICL. The 194 bp Sall- SgrAI fragment was isolated from pICL, and the cohesive ends were converted to blunt ends using E.coli DNA polymerase I (Klenow fragment) and dNTP's. The Asp718 cohesive ends were converted to blunt ends by treatment with mungbean nuclease. By ligation clones were generated that contain the ITR in the Asp718 site of plasmid pICL. A clone that contained the ITR fragment in the correct orientation was designated pICLI (Fig. 18).
Generation of adenovirus Ad-CMV-hcTK. Recombinant
adenovirus was constructed according to the method described in Patent application 95202213. Two components are required to generate a recombinant adenovirus. First, an adaptor-plasmid containing the left terminus of the adenovirus genome containing the ITR and the packaging signal, an expression cassette with the gene of interest, and a portion of the adenovirus genome which can be used for homologous recombination. In addition, adenovirus DNA is needed for recombination with the aforementioned adaptor plasmid In the case of Ad-CMV-hcTK, the plasmid PCMV.TK was used as a basis This plasmid contains nt 1-455 of the adenovirus type 5 genome, nt. 456-1204 derived from pCMVβ (Clontech, the Pstl-StuI fragment that contains the CMV enhancer promoter and the 16S/19S intron from Simian Virus 40), the Herpes Simplex Virus thymidine kmase gene (described in Patent application 95202213), the SV40-derιved polyaoenylation signal (nt 2533-2668 of the SV40 sequence), followed by the Bglll-Scal fragment of Ad5 (nt. 3328-6092 of the Ad5 sequence). These fragments are present in a pMLPIO -derived (Levrero et al., 1991) backbone. To generate plasmid pAD-CMVhc-TK, piasmid pCMV.TK was digested with Clal (the unique Clal-site is located just upstream of the TK open readmgframe) and dephosphorylated with Calf-Intestine Alkaline Phosphate. To generate a hairpin-structure, the synthetic
oligonucleotides HP/cla2 and HP/cla2 were annealed and phopsphorylated on their 5'-OH groups with T4- polynucleotide kinase and ATP. The double-stranded oligonucleotide was ligated with the linearized vector fragment and used to transform E. coli strain "Sure".
Insetion of the oligonucleotide into the Clal site will disrupt the Clal recognition sites. In the oligonucleotide contains a new Clal site near one of its termini. In selected clones, the orientation and the megrity of the inserted oligonucleotide was verified by sequence
analyses. A clone containing the oligonucleotide in the correct orientation (the Clal site at the ITR side) was denoted pAd-CMV-hcTK. This plasmid was co-transfected with Clal digested wild-type Adenovirus-typeS DNA into 911 cells. A recombinant adenovirus in wnich the CMV-hcTK expression cassette replaces the E1 sequences was isolated and propagated using standard procedures.
To study whether the hairpin can be used as a primer for reverse strand synthesis on the displaced strand after replication had started at the ITR, the plasmid pICLhac is introduced into 911 cells (human embryonic retinoblasts transformed with the adenovirus E1 region). The plasmid pICLhaw serves as a control, which contains the
oligonucleotide pair HP/asp 1 and 2 in the reverse orientation but is further completely identical to plasmid pICLhac Also included in these studies are plasmids pICLI and pICL In the plasmid pICLI the hairpin is replaced by an adenovirus ITR Plasmid pICL contains neither a hairpin nor an ITR sequence. These plasmids serve as controls to determine the efficiency of replication by virtue of the terminal-hairpin structure. To provide the viral products other than the E1 proteins (these are produced by the 911 cells) required for DNA replication the cultures are infected with the virus IG.Ad.MLPI.TK after transfection. Several parameters are being studied to demonstrate proper replication of the transfected DNA molecules First, DNA extracted from the cell cultures transfected with
aforementioned plasmids and infected with IG.Ad.MLPI.TK virus is being analyzed by Southern blotting for the presence of the expected replication intermediates, as well as for the presence of the duplicated genomes.
Furthermore, from the transfected and IG.Ad.MLPI.TK infected cell populations virus is isolated, that is capable to transfer and express a luciferase marker gene into luciferase negative cells.
Plasmid DNA of plasmids pICLhac, pICLhaw, pICLI and pICL have been digested with restriction endonuclease Sall and treated with mungbean nuclease to remove the 4 nucleotide single-stranded extension of the resulting DNA fragment. In this manner a natural adenovirus 5' ITR terminus on the DNA fragment is created. Subsequently, both tne pICLhac and pICLhaw plasmids were digested with restriction endonuclease Asp718 to generate tne terminus capable of forming a hairpin structure. The digested plasmids are introduced into 911 cells, using the standard calcium phosphate co-precipitation technique, four dishes for each plasmid. During the transfection, for each plasmid two of the cultures are infected with the
IG.Ad.MLPI.TK virus using 5 infectious IG.Ad.MLPI.TK particles per cell. At twenty-hours post-transfection and fort hours post-transfection one Ad. tk-virus-mfected and one unmfected culture are used to isolate small
molecuiar-weignt DNA using the procedure devised by Hirt. Aliquots of isolated DNA are used for Southern analysis. After digestion of the samples with restriction
endonuclease EcoRI using the luciferase gene as a probe a hybridizing fragment of approx. 2.6kb is detected only in the samples from the adenovirus infected cells transfected with plasmid pICLhac. The size of this fragment is consistent with the anticipated duplication of the luciferase marker gene. This supports the conclusions that the inserted hairpin is capable to serve as a primer for reverse strand synthesis. The hybridizing fragment is absent if the IG.Ad.MLPI.TK virus is omitted, or if the hairpin oligonucleotide has been inserted in the reverse orientation. The restriction endonuclease Dpnl recognizes the tetranucleotide sequence 5'-GATC-3', but cleaves only methylated DNA, (that is, only (plasmid) DNA propagated in, and derived, from E.coli, not DNA that has been replicated m mammalian cells). The restriction
endonuclease Mbol recognizes the same sequences, but cleaves only unmethylated DNA (viz. DNA propagated m mammalian cells). DNA samples isolated from the
transfected cells are incubated with Mbol and Dpnl and analysed with Southern blots. These results demonstrate that only in the cells transfected with the pICLhac and the pICLI plasmids large Dpnl-resistant fragments are present, that are absent in the Mbol treated samples These data demonstrate that only after transfection of plasmids pICLI and pICLhac replication and duplication of the fragments occur.
These data demonstrate that in -adenovirus-infected cells linear DNA fragments that have on one terminus an adenovirus-derived inverted terminal repeat (ITR) and at the other terminus a nucleotide sequence that can anneal to sequences on the same strand, when present in single- stranded form thereby generate a hairpin structure, and will be converted to structures that have inverted terminal repeat sequences on both ends The resulting DNA molecules will replicate by the same mechanism as the wild type adenovirus genomes.
1.2 Demonstration that the DNA molecules that contain a luciferase marker gene, a single copy of the ITR, the encapsidation signal and a synthetic DNA
sequence, that is capable of forming a hairpin structure, are sufficient to generate DNA molecules that can be encapsidated into virions.
To demonstrate that the above DNA molecules
containing two copies of the CMV-luc marker gene can be encapsidated into virions, virus is harvested from the remaining two cultures via three cycles of freeze-thaw crushing and is used to infect murme fibroblasts. Fortyeight hours after infection the infected cells are assayed for luciferase activity. To exclude the possibility that the luciferase activity has been induced by transfer of free DNA, rather than via virus particles, virus stocks are treated with DNasel to remove DNA contaminants
Furthermore, as an additional control, aliquots of the virus stocks are incubated for 60 minutes at 56°C The neat treatment will not affect the contaminating DNA, but will inactivate the viruses. Significant luciferase activity is only found in the cells after infection with the virus stocks derived from IG.Ad.MLPI.TK-infected cells
transfected with the pICLhc and pICLI plasmids Neither in the non-infected cells, nor in the infected cells
transfected with the pICLhw and pICL significant
luciferase activity can be demonstrated. Heat
mactivation, but not DNasel treatment, completely
eliminates luciferase expression, demonstrating that adenovirus particles and not free (contaminating) DNA fragments are responsible for transfer of the luciferase reporter gene.
These results demonstrate that these small viral genomes can be encapsidated into adenovirus particles and suggest that the ITR and the encapsidation signal are sufficient for encapsidation of linear DNA fragments into adenovirus particles. These adenovirus particles can be used for efficient gene transfer. When introduced into cells that contain ano express at least part of the adenovirus genes (viz. E1, E2, E4, and L, and VA),
recombinant DNA molecules that consist of at least one
ITR, at least part of the encapsidation signal as well as a synthetic DNA sequence, that is capable of forming a hairpin structure, have the intrinsic capacity to
autonomously generate recombinant genomes which can be encapsidated into virions Such genomes and vector system can be used for gene transfer. 1.3 Demonstration that DNA molecules which contain nucleotides 3510 - 35953 (viz. 9.7 - 100 map units) of the adenovirus type 5 genome (thus lack the E1 protein-coding regions, the right-hand ITR and the encapsidation
sequences) and a terminal DNA sequence that is
complementary to a portion of the same strand of the DNA molecule when present in single-stranded form other than the ITR, and as a result is capable of forming a hairpin structure, can replicate in 911 cells.
In order to develop a replicating DNA molecule that can provide the adenovirus products required to allow the above mentioned ICLhac vector genome and alike minimal adenovectors to be encapsidated into adenovirus particles by nelper cells, the Ad-CMV-hcTK adenoviral vector has been developed. Between the CMV enhancer/promoter region and the thvmidme kmase gene the annealed oligonucleotide pair HP/cla 1 and 2 is inserted. The vector Ad-CMV-hcTK can be propagated and produced in 911 cell using standard procedures. This vector is grown and propagated
exclusively as a source of DNA used fcr transfection. DNA of the adenovirus Ad-CMV-hcTK is isolated from virus particles that had been purified using CsCl density- gradient centnfugation by standard techniques. The virus DNA has been digested with restriction endonuclease Clal. The digested DNA is s ize- fractionated on an 0.7% agarose gel and the large fragment is isolated and used for further experiments. Cultures of 911 cells are transfected large Clal-fragment of the Ad-CMV-hcTK DNA using the standard calcium phosphate co-precipitation technique. Much like in the previous experiments with plasmid pICLhac, the AD-CMV-hc will replicate starting at the right-hand ITR. Once the 1-strand is displaced, a hairpin can be formed at the left-hand terminus of the fragment. This facilitates the DNA polymerase to elongate the chain towards the rignt-hand-side. The process will proceed until the displaced strand is completely converted to its double-stranded form. Finally, the right-nand ITR will be recreated, and in this location the normal adenovirus replication-initiation and elongation will occur. Note that the polymerase will read through the hairpin, thereby duplicating the molecule. The input DNA molecule of 33250 bp, that had on one side an adenovirus ITR sequence and at the other side a DNA sequence that had the capacity to form a nairpm structure, has now been duplicated, in a way that both ends contain an ITR sequence. The resulting DNA molecule will consist of a palindromic structure of approximately 66500 bp.
This structure can be detected m low-molecular weight DNA extracted from the transfected cells using Southern analysis. The palindromic nature of the DNA fragment can be demonstrated by digestion of the low- molecular weight DNA with suitable restriction
endonucleases and Southern blotting with the HSV-TK gene as the probe. This molecule can replicate itself in the transfected cells by virtue of the aoenovirus gene products that are present in the cells. In part, the adenovirus genes are expressed from templates that are integrated in the genome of the target cells (viz. the E1 gene products), the other genes reside in the replicating DNA fragment itself. Note however, that this linear DNA fragment cannot be encapsidated into virions. Not only does it lack all the DNA sequences required for
encapsidation, but also is its size much too large to be endapsioated.
1.4 Demonstration that DNA molecules which contain nucleotides 3503 - 35953 (viz. 9.7 - 100 map units) of the adenovirus type 5 genome (thus lacκ the E1 protein-coding regions, the right-hand ITR and the encapsidation
sequences) and a terminal DNA sequence tnat is
complementary to a portion the same strand of the DNA molecule other than the ITR, and as a result is capable of forming a hairpin structure, can replicate in 911 cells and can provide the helper functions required to
encapsioate the pICLI and pICLhac derived DNA fragments. The next series of experiments aim to demonstrate that the DNA molecule described in part 1.3 could be used to encapsidate the minimal adenovectors described in part 1.1 and 1.2.
In the experiments the large fragment isolated after endonuclease Clal-digestion of Ad-CMV-hcTK DNA is
introduced into 911 cells (conform the experiments described m part 1.3) together with endonuclease Sall, mungbean nuclease, endonuclease Asp718-treated plasmid pICLhac, or as a control similarly treated plasmid pICLhaw. After 48 hours virus is isolated by freeze-thaw crushing of the transfected cell population. The virus- preparation is treated with DNasel to remove contaminating free DNA. The virus is used subsequently to infect Rat2 fibroblasts. Forty-eight hours post infection the cells are assayed for luciferase activity Only in tne cells infected with virus isolated from the cells transfected with the pICLhac plasmid, and not with the pICLhaw plasmid, significant luciferase activity can oe
demonstrated. Heatinactivation of the virus prior to infection completely abolishes the luciferase activity, indicating that the luciferase gene is transferred by a "iral particle. Infection of 911 cell with the virus stock did not result in any cytopathological effects,
demonstrating that the pICLhac is produced without any infectious helper virus that can be propagated on 911 cells. These results demonstrate that the proposed method can be used to produce stocks of minimal-adenoviral vectors, that are completely devoid of infectious helper viruses that are able to replicate autonomously on adenovirus-transformed human cells or on non-adenovirus transformed human cells.
Besides the system described in this application, another approach for the generation of minimal adenovirus vectors has been disclosed in WO 94/12649. The method described in WO 94/12649 exploits the function of the protein IX for the packaging of minimal adenovirus vectors (Pseudo Adenoviral Vectors (PAV) in the terminology of WO 94/12649). PAVs are produced by cloning an expression plasmid with the gene of interest between the left-hand (including the sequences required for encapsidation) and the right-hand adenoviral ITRs. The PAV is propagated in the presence of a helper virus. Encapsidation of the PAV is preferred compared the helper virus because the helper virus is partially defective for packaging. (Either by virtue of mutations in the packaging signal or by virtue of its size (virus genomes greater than 37.5 kb package inefficiently). In addition, the authors propose that in the absence of the protein IX gene the PAV will be preferentially packaged. However, neither of these mechanisms appear to be sufficiently restrictive to allow packaging of only PAVs/mmimal vectors. The mutations proposed in the packaging signal dimmish packaging, but do not provide an absolute block as the same packagingactivity is required to propagate the helper virus Also neither an increase in the size of the helper virus nor the mutation of the protein IX gene will ensure that PAV is packaged exclusively. Thus, the method described in WO 94/12649 is unlikely to be useful for the production of helper-free stocks of minimal adenovirus vectors/PAVs.
References
Berk, A. J. (1986): Ann . Rev. genet . 20, 45-79.
Bernards, R., Schrier, P. I., Bos, J.L., and Eb,
A.J. v. d. (1983): Role of adenovirus types 5 and 12 early region lb tumor antigens in oncogenic
transformation. Virology 127, 45-53.
Bett, A.J., Prevec, L., and Graham, F.L. (1993): Packaging Capacity and Stability of Human Adenovirus Type-5 Vectors. J Virol 67, 5911-5921.
Blaese, M., Blankenstein, T., Brenner, M., Cohen- Haguenauer, O., Gansbacher, B., Russell, S.,
Sorrentmo, B., and Velu, T. (1995). Vectors in cancer therapy: how will they deliver? Cancer Gene Ther . 2 , 291-297.
Boshart, M., Weber, F., Jahn, G., Dorsch-Häler, K.,
Fleckenstein, B., and Scaffner, W. (1985): A very strong enhancer is located upstream of an immediate early gene of human Cytomegalovirus. Cell 41,
521-530.
Bout, A., Imler, J.L., Schulz, H., Perricaudet, M.,
Zurcher, C., Herbrink, P., Valerio, D., and Pavirani, A. (1994a): In vivo adenovirus-mediated transfer of human CFTR cDNA to Rhesus monkey airway epithelium: efficacy, toxicity and safety Gene Therapy 1,
385-394.
Bout, A., Pemcaudet, M., Baskm, G., Imler, J. L.,
Scholte, B.J., Pavirani, A., and Valerio, D. (1994b): Lung gene therapy: in vivo adenovirus mediated gene transfer to rhesus monkey airway epithelium.
Human Gene Therapy 5, 3-10.
Brody, S.L., and Crystal, R.G. (1994) : Adenovirus-mediated in vivo gene transfer. Ann N Y Acad Scl 716 , 90-101. Brough, D.E., Cleghon, V., and Klessig, D.F. (1992).
Construction, characterization, and utilization of cell lines which mducibly express the adenovirus DNA-binding protein. Virology 190 (2), 624-34. Brough, D.E., Rice, S.A., Sell, S., and Klessig, D.F.
(1985): Restricted changes in the adenovirus
DNA-bmdmg protein that lead to extended host range or temperature-sensitive phenotypes. J . Virol . 55, 206-212.
Daniell, E. (1976): Genome structure of incomplete
particles of adenovirus. J Virol . 19, 685-708.
E1sen, P.J.V. d., Houwelmg, A., and Eb, A. J.V. d.
(1983). Expression of region E1B of human adenoviruses in tne absence of region E1A is not sufficient for complete transformation. Virology 128, 377-390.
Engelhardt, J.F., Litzky, L., and Wilson, J.M. (1994a)
Prolonged transgene expression in cotton rat lung with recombinant adenoviruses defective in E2A.
Hum . Gene Ther . 5, 1217-1229.
Engelhardt, J.F., Simon, R.H., Yang, Y., Zepeda, M.,
Weber-Pendleton, S., Doranz, B., Grossman, M., and Wilson, J.M. (1993). Adenovirus-mediated transfer of the CFTR gene to lung of nonnuman primates : biological efficacy study. Human Gene Therapy 4, 759-769.
Engelhardt, J.F., Ye, X., Doranz, B. , and Wilson, J.M.
(1994b): Ablation of E2A in recombinant adenoviruses improves transgene persistence and decreases
inflammatory response in mouse liver. Proc Na tl Acad Scl U S A 91, 6196-200.
Fang, B., Wang, H., Gordon, G., Bellinger, D.A.,
Read, M.S., Brinkhous, K.M., Woo, S.L.C., and
Eisensmith, R.C. (1996). Lack of persistence of E1- recombinant adenoviral vectors containing a
temperature sensitive E2A mutation in immunocompetent mice and hemophilia dogs. Gene Ther . 3, 217-222.
Fallaux F.J., Kranenburg, O., Cramer, S.J.,
Houweling, A., Ormondt, H. v., Hoeben, R.C. , and Eb, A.J v.d. (1996). Characterization of 911. a new helper cell line for the titration and propagation of early-region- 1-deleted adenoviral vectors.
Hum. Gene Ther. 7, 215-222. Gooding, L.R., Aquino, L., Duerksen-Hughes, P.J., Day, D., Horton, T.M., Yei, S., and Wold, W.S.M. (1991): The E1B 19, 000-molecular-weight protein of group C adenoviruses prevents tumor necrosis factor cytolysis of human cells but not of mouse cells. J. Virol . 65, 3083-3094.
Gräble, M., and Hearing, P. (1990): Adenovirus type 5
packaging domain is composed of a repeated element that is functionally redundant. J. Virol . 64,
2047-2056.
Gräble, M., and Hearing, P. (1992): cis and trans
Requirements for the Selective Packaging of Adenovirus Type-5 DNA. J Virol 66, 723-731.
Graham, F.L., and van der Eb, A.J. (1973). A new technique for the assay of infectivity of human adenovirus 5 DNA. Virology 52, 456 - 467.
Graham, F.L., Smiley, J., Russell, W.C., and Nairn, R.
(1977): Characteristics of a human cell line
transformed by DNA from adenovirus type 5
J. Gen . Virol . 36, 59-72.
Haddada, H., Ragot, T., Cordier, L., Duffour, M.T., and
Perricaudet, M. (1993): Adenoviral mterleukm-2 gene transfer into P815 tumor cells abrogates
tumorigenicity and induces antitumoral immunity in mice. Hum Gene Ther 4, 703-11.
Hay, R.T., Stow, N.D., and McDougall, I.M. (1984).
Replication of adenovirus mmichromosomes. J. Mol . Biol . 174, 493-510.
Hearing, P., Samulski, R.J., Wishart, W.L., and Shenk, T.
(1987): Identification of a repeated sequence element required for efficient encapsidation of the adenovirus type 5 chromosome. J. Virol . 61, 2555-2558.
Horwitz, M.S. (1990). Adenoviridae and their replication, pp. 1679-1740. In B.N. Fields, and D.M. Knipe (Eds): Virology, Raven Press, Ltd, New York.
Hu, C.H., Xu, F.Y., Wang, K., Pearson, A. N., and Pearson, G. D. (1992): Symmetrical Adenovirus Mmichromosomes Have Hairpin Replication Intermediates. Gene 110, 145-150.
Imler, J.L., Chartier, C., Dreyer, D., Dieterle, A.,
Sainte-Marie, M., Faure, T., Pavirani, A., and
Mehtali, M. (1996). Novel complementation cell lines derived from human lung carcinoma A549 cells support the growth of E1 -deleted adenovirus vectors.
Gene Ther . 3, 75-84
Jochemsen, A.G., Peltenburg, L.T.C., Pas, M.F.W.T.,
Wit, CM. d., Bos, J.L., and Eb, A.J. v.d. (1987) : EMBO J. 6, 3399-3405.
Klessig, D.F., and Grodzicker, T. (1979): Mutations that allow human Ad2 and Ad5 to express late genes in monxey cells maps in the viral gene encoding the "2K DNA-binding protein. Cell 17, 957-966.
Klessig, D.F., Grodzicker, T., and Cleghon, V. (1984):
Construction of human cell lines which contain and express the adenovirus DNA binding protein gene by cotransformation with the HSV-1 tk gene. Virus Res . 1, 169-188.
Kruijer, W., Nicolas, J.C., Schaik, F.M. v., and
Sussenbach, J.S. (1983): Structure and function of DNA bmomg proteins from revertants of adenovirus type 5 mutants with a temperature-sensitive DNA replication. Virology 124, 425-433
Lechner, R.L., and Kelly Jr., T.J. (1977). The structure of replicating adenovirus 2 DNA molecules. J. Mol . Biol . 174, 493-510.
Leij, L. de, Postmus, P. E., Buys, C.H.C.M., E1ema, J.D., Ramaekers, F., Poppema, S., Brouwer, M. , Veen, A.Y. v.d., Mesander, G., and The, T.H. (1985):
Characterization of three new variant type cell lines derived from small cell carcinoma of the lung.
Cancer Res . 45, 6024-6033.
Levrero, M., Barban, V., Manteca, S., Ballay, A., Balsamo, C. , Avantaggiati, M.L., Natoli, G., Skellekens, H., Tiollais, P. , and Perricaudet, M. (1991) : Defective and nondefective adenovirus vectors for expressing foreign genes in vitro and in vivo . Gene 101,
195-202.
Lochmuller, H., Jani, A., Huard, J., Prescott, S.,
Simoneau, M., Massie, B., Karpati, G., and Acsadi, G. (1994): Emergence of early region 1-contamιng replication-competent adenovirus in stocks of
replication-defective adenovirus recombmants
(ΔE1 + ΔE3 ) during multiple passages in 293 cells. Hum . Gene Ther . 5, 1485-1492.
Matsui T, Murayama M. and Mita T (1986) Adenovirus 2
peptide IX is expressed only on replicated DNA molecules. Mol . Cell Biol . 6, 4149-4154.
Michelson, A.M., Markham, A.F. , and Orkin, S.H. (1983) :
Isolation and DNA sequence of a full-length cDNA clone for human X-chromosome encoded phosphoglycerate kmase. Proc Na tl . Acad . Sci . USA 80, 472-476.
Morm, J.E., Lubeck, M.D., Barton, J. E., Conley, A.J., Davis, A.R., and Hung, P.P. (1987): Recombinant adenovirus induces antibody reponse to hepatitis B virus surface antigens. Proc . Natl . Acad . Sci . USA 84, 4626-4630.
Nicolas, J.C., Suarez, F., Levine, A.J. , and Girard, M.
(1981) : Temperature-independent revertants of
adenovirus H5ts125 and H5ts107 mutants m the DNA binding protein: isolation of a new class of host range temperature conditional revertants. Virology 108, 521-524.
Ostrove, J.M. (1994): Safety testing programs for gene
therapy viral vectors. Cancer Gene Ther . 1, 125-131. Pacmi, D.L., Dubovi, E.J., and Clyde, W.A. (1984) :
J. Infect . Dis . 150, 92-97
Postmus, P.E., Ley, L.d., Veen, A.Y. v d., Mesander, G.,
Buys, C.H.C.M ., and E1ema, J.D. (1988) Two small cell lung cancer cell lines established from rigid
bronchoscope biopsies. Eur. J. Cl in. Oncol . 24,
753-763. Rice, S.A., and Klessig, D.F. (1985): Isolation and analysis of adenovirus type 5 mutants containing deletions in the gene encoding tne DNA-binding
protein. J. Virol . 56, 767-778.
Roberts, B.E., Miller, J.S., Kimelman, D., Cepko, C.L.,
Lemischka, I. R., and Mulligan, R. C. (1985):
J. Virol . 56, 404-413.
Shapiro, D.L., Nardone, L.L., Rooney, S.A., Motoyama,
E.K., and Munoz, J.L. (1978) Phospnoiipid
biosynthesis and secretion by a cell line (A549) which resembles type II alveolar epithelial cells. Biochim .
Biophys . Acta 530, 197-207
Simon, R. H., Engelhardt, J.F., Yang Y., Zepeda, M.,
Weoer-Pendleton, S., Grossman, M., and Wilson, J.M. (1993) : Adenovirus-mediated transfer of the CFTR gene to lung of nonhuman primates: toxicity study.
Human Gene Therapy 4, 771-780
Singer-Sam, J., Keith, D.H., Tani, K. , Simmer, R.L.,
Shively, L., Lindsay, S, Yoshica, A., and Riggs, A.D. (1984): Sequence of the promoter region of the gene for X-linked 3-phosphoglycerate kinase. Gene 32,
409-417.
Stem, R.W. , and Whelan, J. (1989) Insulin gene enhancer activity is inhibited by adenovirus 5 E1A gene
products. Mol Cell Biol 9, 4531-4.
Stratford-Pemcaudet, L.D., and Perricaudet, M. (1991):
Gene transfer into animals, the promise of adenovirus, pp 51-61. In 0. Cohen-Adenauer and M. Boiron (Eds):
Human Gene Transfer, John Libbey Eurotext.
Telling, G.C., Perera, S., Szalkowsxi, O.M., and Williams,
J. (1994): Absence of an essential regulatory
influence of the adenovirus E1B 19-kιlodalton protein on viral growth and early gene expression in human diploid WI38, HeLa, and A549 cells. J. Virol 68,
541-7.
Tooze, J. (1981): DNA Tumor Viruses (revised) . Cold Spring
Harbor Laboratory. Cold Spring Harbor, New York. Vieira, J., and Messing, J. (1987): Production of single stranded plasmid DNA, pp. 3-11: Methods in Enzymology, Acad. Press Inc.
Vincent, A.J.P.E., Esandi, M. d. C, Someren,
G.D. v., Noteboom, J.L., C.J.J, A., Vecht, C.,
Smitt, P.A.E.S., Bekkum, D.W. v., Valerio, D.,
Hoogerbrugge, P.M., and Bout, A. (1996a). Treatment of Lepto-menmgeal metastasis in a rat model using a recombinant adenovirus containing the HSV-tk gene. J . Neurosurg . in press.
Vincent, A.J.P.E., Vogels, R., Someren, G. v.,
Esandi, M. d. C, Noteboom, J.L., Avezaat, C.J.J., Vecht, C., Bekkum, D.W. v., Valerio, D., Bout, A., and Hoogerbrugge, P.M. (1996b). Herpes Simplex Virus Thymidine Kmase gene therapy for rat malignant brain tumors. Hum . Gene Ther . 7, 197-205.
Wang, K., and Pearson, G.D. (1985): Adenovirus sequences required for replication in vivo . Nucl . A cids Res . 13, 5173-5187.
White, E., Denton, A., and Stillman, B. (1988): J. Virol .
62, 3445-3454.
Yang, Y., Li, Q., Ertl, H.C.J., and Wilson, J.M. (1995):
Cellular and humoral immune responses to viral antigens create barriers to lung-directed gene therapy with recombinant adenoviruses. J Virol . 69,
2004-2015.
Yang, Y., Nunes, F.A., Berencsi, K., Furth, E.E., Gonczol, E., and Wilson, J.M. (1994a): Cellular immunity to viral antigens limits E1-deleted adenoviruses for gene therapy. Proc Natl Acad Sci U S A 91, 4407-11.
Yang, Y . , Nunes, F. A., Berencsi, K., Gonczol, E.,
Engelhardt, J.F., and Wilson, J. M. (1994b):
Inactivation of E2A in recombinant adenoviruses improves the prospect for gene therapy in cystic fibrosis. Nat Genet 7, 362-9. Zantema, A., Fransen, J.A.M., Davis-Olivier, A.,
Ramaekers, F.C.S., Vooijs, G.P., Deleys, B., and Eb, A.J. v.d. (1985). Localization of the E1B proteins of adenovirus 5 in transformed cells, as revealed by interaction with monoclonal antibodies. Virology 142, 44-58.
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001

Claims

1. A recombinant nucleic acid molecule based on or derived from an adenovirus having at least a functional encapsidatmg signal and at least one functional Inverted Terminal Repeat or a functional fragment or derivative thereof and having no overlapping sequences which allow for homologous recombination leading to replication competent virus in a cell into which it is transferred.
2. A recombinant nucleic acid molecule according to claim 1 being in a linear form and comprising an Inverted Terminal Repeat at or near both termini.
3. A recombinant nucleic acid molecule according to claim 1 being in a linear and essentially single stranded form and comprising at the 3' terminus a sequence
complementary to an upstream part of the same strand of said nucleic acid molecule, said sequence being capable of base-pairing with said part in a way to be able to function as a start-site for a nucleic acid polymerase.
4. A recombinant nucleic acid molecule according to claim 3, comprising all adenovirus derived genetic information necessary for replication, except for a functional encapsidation signal.
5. A recombinant nucleic acid molecule derived from tne nucleic acid molecule according to claim 4 resulting from the action of a nucleic acid polymerase on said nucleic acid molecule according to claim 4.
6. A recombinant nucleic acid molecule according to claim 5 having an Inverted Terminal Repeat at both termini.
7. A recombinant nucleic acid molecule according to anyone of the aforegoing claims comprising a host range mutation.
8. A recombinant nucleic acid molecule according to anyone of the aforegoing claims comprising a mutated E2 region rendering at least one of its products temperature sensitive.
9. A recombinant nucleic acid molecule according to anyone of the aforegoing claims comprising an E2 region under the control of an inducible promoter.
10. A packaging cell for packaging adenovirus derived nucleic acid molecules, which packaging cell has been provided with one or more recombinant nucleic acid molecules which provide said cell with the aoility to express adenoviral gene products derived from at least the E1A region.
11. A packaging cell for packaging adenovirus derived nucleic acid molecules, which packaging cell has been provided with one or more recombinant nucleic acid molecules which provide said cell with the ability to express adenoviral gene products derived from at least both the E1A and the E2A region.
12. A packaging cell according to claim 11, wherein the recompinant nucleic acid molecule encoding the E2A region is under control of an mducible promoter.
13. A packaging cell according to claim 11 or 12, wherein the recombinant nucleic acid molecule encoding the E2A region is mutated so that at least one of its products is temperature sensitive.
14. A cell according to anyone of claims 10-13, which does not have the aoility to express E1B products.
15. A cell according to claim 14, wherein the genetic information encoding E1B products is not present.
16. A cell according to claim 10, further comprising the region coding for E1B.
17. A cell according to claim 10, further comprising a marker gene.
18. A cell according to claim 17, whereby the marker gene is under control of the E1B responsive promoter.
19. A packaging cell harbouring nucleotides 80-5788 of the human Adenovirus 5 genome.
20. A packaging cell harbouring nucleotides 459-1713 of the human Adenovirus 5 genome.
21. A packaging cell harbouring nucleotides 459-3510 of the human Adenovirus 5 genome.
22. A ceil according to anyone of claims 10-13, which does not have the ability to express hne 21kD E1B product.
23. A cell according to claim 22, wherein the genetic information encoding the 21kD E1B product is not present
24. A cell according to anyone of claims 10-23 which is a diploid cell.
25. A cell according to anyone of claims 10-24 which is of non-human origin.
26. A cell according to anyone of claims 10-25 which is of monkey origin.
27. A cell according to claim 19 as deposited under no. 95062101 at the ECACC.
28. A recombinant nucleic acid molecule according to anyone of claims 1-9 being a DNA molecule.
29. A recomoinant nucleic acid molecule based on or derived from an adenovirus, having at least a deletion of nucleotides 459-3510 of the E1 region.
30. A recomoinant nucleic acid molecule based on or derived from an adenovirus, having a deletion of
nucleotides 459-1713 of the E1 region.
31. An adenovirus-like particle comprising a recombinant nucleic acid molecule according to anyone of claims 1-9.
32. A cell comprising a recombinant nucleic acid molecule according to anyone of claims 1-9.
33. A recombinant nucleic acid according to claims 1-3, comprising functional E2A end E2B genes or functional fragments or derivatives thereof under control of an E1A independent promoter.
34. A cell according to claim 26 which comprises a host range mutated E2A region of an adenovirus.
35. A method for intracellular amplification comprising the steps of providing a cell with a linear DNA fragment to be amplified, which fragment is provided with at least a functional part or derivative of an Inverted Terminal Repeat at one terminus and providing said cell with functional E2 derived products necessary for replication of said fragment and allowing said fragment to be acted upon by a DNA polymerase.
36. A method according to claim 35 whereby the cell is provided with genetic material encoding both e2A and E2B products.
37. A method according to claim 35 or 36 wnereby a hairpin-like structure is provided at the terminus of the DNA fragment opposite the Inverted Terminal Repeat.
PCT/NL1996/000244 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy WO1997000326A1 (en)

Priority Applications (26)

Application Number Priority Date Filing Date Title
JP50294897A JP4051416B2 (en) 1995-06-15 1996-06-14 Packaging system for human recombinant adenovirus used in gene therapy
DK96917735.1T DK0833934T4 (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus for use in gene therapy
IL16040696A IL160406A0 (en) 1995-06-15 1996-06-14 A cell harbouring nucleic acid encoding adenoritus e1a and e1b gene products
DK04101716.1T DK1445322T4 (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus for use in gene therapy
DE69633565T DE69633565T3 (en) 1995-06-15 1996-06-14 PACKAGING SYSTEMS FOR HUMAN, HUMAN ADENOVIRES, FOR USE IN GENE THERAPY
EP96917735A EP0833934B2 (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy
SI9630698T SI0833934T2 (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy
CA2222140A CA2222140C (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy
KR10-2004-7010409A KR100470180B1 (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy
US08/793,170 US5994128A (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy
IL12261496A IL122614A0 (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy
AT96917735T ATE278794T1 (en) 1995-06-15 1996-06-14 PACKAGING SYSTEMS FOR HUMAN, HUMAN ADENOVIRUSES, FOR USE IN GENE THERAPY
ES96917735T ES2231813T5 (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus used in gene therapy
AU60182/96A AU731767B2 (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy
US09/333,820 US6306652B1 (en) 1995-06-15 1999-06-15 Packaging systems for human recombinant adenovirus to be used in gene therapy
US09/918,029 US6783980B2 (en) 1995-06-15 2001-07-30 Packaging systems for human recombinant adenovirus to be used in gene therapy
US10/038,271 US20020151032A1 (en) 1995-06-15 2001-10-23 Packaging systems for human recombinant adenovirus to be used in gene therapy
US10/125,751 US7105346B2 (en) 1995-06-15 2002-04-18 Packaging systems for human recombinant adenovirus to be used in gene therapy
US10/219,414 US20030104626A1 (en) 1995-06-15 2002-08-15 Packaging systems for human recombinant adenovirus to be used in gene therapy
US10/618,526 US20050260596A1 (en) 1995-06-15 2003-07-11 Packaging systems for human recombinant adenovirus to be used in gene therapy
IL160406A IL160406A (en) 1995-06-15 2004-02-15 Cell harbouring nucleic acid encoding adenovirus e1a and e1b gene products
US10/850,140 US7052881B2 (en) 1995-06-15 2004-05-20 Packaging systems for human recombinant adenovirus to be used in gene therapy
US11/134,674 US8236293B2 (en) 1995-06-15 2005-05-19 Means and methods for nucleic acid delivery vehicle design and nucleic acid transfer
US11/485,114 US20060246569A1 (en) 1995-06-15 2006-07-12 Packaging systems for human recombinant adenovirus to be used in gene therapy
US11/879,421 US20090023196A1 (en) 1995-06-15 2007-07-16 Stocks of replication-deficient adenovirus
US11/900,463 US20080138901A1 (en) 1995-06-15 2007-09-11 Packaging systems for human recombinant adenovirus to be used in gene therapy

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP95201611 1995-06-15
EP95201611.1 1995-06-15
EP95201728 1995-06-26
EP95201728.3 1995-06-26

Related Child Applications (6)

Application Number Title Priority Date Filing Date
US08793170 A-371-Of-International 1996-06-14
US08/793,170 A-371-Of-International US5994128A (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy
US08/793,170 Continuation US5994128A (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy
US09/298,745 Continuation-In-Part US6395519B1 (en) 1995-06-15 1999-04-23 Means and methods for nucleic acid delivery vehicle design and nucleic acid transfer
US09/334,765 Continuation US6238893B1 (en) 1995-06-15 1999-06-16 Method for intracellular DNA amplification
US09/356,575 Continuation-In-Part US6265212B1 (en) 1995-06-15 1999-07-19 Packaging systems for human recombinant adenovirus to be used in gene therapy

Publications (1)

Publication Number Publication Date
WO1997000326A1 true WO1997000326A1 (en) 1997-01-03

Family

ID=26139401

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL1996/000244 WO1997000326A1 (en) 1995-06-15 1996-06-14 Packaging systems for human recombinant adenovirus to be used in gene therapy

Country Status (15)

Country Link
US (10) US5994128A (en)
EP (2) EP0833934B2 (en)
JP (2) JP4051416B2 (en)
KR (2) KR100449181B1 (en)
AT (2) ATE445705T1 (en)
AU (1) AU731767B2 (en)
CA (1) CA2222140C (en)
DE (2) DE69633565T3 (en)
DK (2) DK0833934T4 (en)
ES (2) ES2231813T5 (en)
HK (1) HK1064406A1 (en)
IL (3) IL122614A0 (en)
PT (2) PT1445322E (en)
SI (2) SI1445322T2 (en)
WO (1) WO1997000326A1 (en)

Cited By (123)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999025861A2 (en) * 1997-11-17 1999-05-27 Aventis Pharma S.A. Adenovirus vectors and method for reducing homologous recombination phenomena
WO1999027123A2 (en) * 1997-11-26 1999-06-03 Board Of Regents, The University Of Texas System Modified sv40 viral vectors
WO1999032647A1 (en) * 1997-12-23 1999-07-01 Introgene B.V. Adeno-associated virus and adenovirus chimeric recombinant viruses useful for the integration of foreign genetic information into the chromosomal dna of target cells
WO1999055132A2 (en) * 1998-04-24 1999-11-04 Introgene B.V. Generation of packaging system for human recombinant adenoviral vectors
US5981225A (en) * 1998-04-16 1999-11-09 Baylor College Of Medicine Gene transfer vector, recombinant adenovirus particles containing the same, method for producing the same and method of use of the same
WO1999064582A2 (en) * 1998-06-12 1999-12-16 Introgene B.V. High-throughput screening of gene function using libraries for functional genomics applications
EP0973866A1 (en) 1997-03-04 2000-01-26 Baxter International Inc. Adenovirus e1-complementing cell lines
WO2000024916A1 (en) * 1998-10-27 2000-05-04 Crucell Holland B.V. Improved aav vector production
JP2000157289A (en) * 1998-11-20 2000-06-13 Introgene Bv Gene delivery vector provided with tissue tropism to smooth muscle cell and/or endothelial cell
WO2000063403A2 (en) * 1999-04-15 2000-10-26 Crucell Holland B.V. Recombinant protein production in a human cell using sequences encoding adenovirus e1 protein
US6140103A (en) * 1995-12-01 2000-10-31 Introgene B.V. Regulated protein expression in stably transfected mammalian cells
WO2001007571A1 (en) * 1999-04-23 2001-02-01 Introgene B.V. Means and methods for nucleic acid transfer
EP1083229A1 (en) * 1999-09-10 2001-03-14 Introgene B.V. Modified adenoviral vectors for use in gene therapy
EP1083228A1 (en) * 1999-09-10 2001-03-14 Introgene B.V. Modified adenoviral vectors for use in gene therapy
US6204052B1 (en) 1994-08-16 2001-03-20 Introgene B.V. Adenoviral vectors with reduced TNF response and partial E3 region deletion
WO2001020014A1 (en) * 1999-09-10 2001-03-22 Crucell Holland B.V. Modified adenoviral vectors for use in gene therapy
WO2001023597A2 (en) * 1999-09-29 2001-04-05 The Trustees Of The University Of Pennsylvania Cell lines and constructs useful in production of e1-deleted adenoviruses in absence of replication competent adenovirus
WO2001036615A2 (en) * 1999-11-18 2001-05-25 Stefan Kochanek Permanent amniocyte cell line, the production thereof and its use for producing gene transfer vectors
US6261554B1 (en) 1995-07-25 2001-07-17 Introgene B.V. Compositions for targeted gene delivery
US6265212B1 (en) 1995-06-15 2001-07-24 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
US6312957B1 (en) 1996-12-05 2001-11-06 Introgene B.V. Genetic modification of primate hemopoietic repopulating stem cells
WO2002031169A1 (en) * 2000-10-10 2002-04-18 Genvec, Inc. Method of preparing a eukaryotic viral vector
WO2002040665A2 (en) * 2000-11-15 2002-05-23 Crucell Holland B.V. Complementing cell lines
US6395519B1 (en) 1995-06-15 2002-05-28 Introgene B.V. Means and methods for nucleic acid delivery vehicle design and nucleic acid transfer
US6447768B1 (en) * 1998-12-30 2002-09-10 Introgene B.V. Methods of gene therapy with a DNA sequence encoding NOS
US6472212B1 (en) 1991-10-04 2002-10-29 Introgene B.V. Methods and compositions for genetically modifying primate bone marrow cells
JP2002537816A (en) * 1999-03-04 2002-11-12 イントロヘーネ ベスローテン フェンノートシャップ Means and methods for transducing fibroblast-like or macrophage-like cells
US6498027B1 (en) 1998-05-20 2002-12-24 Introgene B.V. Targeted delivery through a cationic amino acid transporter
US6558948B1 (en) 1999-11-23 2003-05-06 Stefan Kochanek Permanent amniocytic cell line, its production and use for the production of gene transfer vectors
AU768489B2 (en) * 1998-11-06 2003-12-11 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Cells for the production of helper dependent adenoviral vectors, method for the preparation and use thereof
US6682929B2 (en) 2001-07-23 2004-01-27 Genvec, Inc. Adenovector complementing cells
US6733993B2 (en) 2000-09-15 2004-05-11 Merck & Co., Inc. Enhanced first generation adenovirus vaccines expressing codon optimized HIV1-gag, pol, nef and modifications
US6787351B2 (en) 1999-07-06 2004-09-07 Merck & Co., Inc. Adenovirus carrying gag gene HIV vaccine
US6803194B1 (en) 1998-02-13 2004-10-12 Hk Pharmaceuticals, Inc. Use of ribozymes for functionating genes
US6855544B1 (en) 1999-04-15 2005-02-15 Crucell Holland B.V. Recombinant protein production in a human cell
US6869936B1 (en) 1999-03-04 2005-03-22 Crucell Holland B.V. Means and methods for fibroblast-like or macrophage-like cell transduction
US6913922B1 (en) 1999-05-18 2005-07-05 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US6929946B1 (en) 1998-11-20 2005-08-16 Crucell Holland B.V. Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
WO2006031689A2 (en) 2004-09-13 2006-03-23 Genzyme Corporation Multimeric constructs
EP1644491A2 (en) * 2003-07-03 2006-04-12 Cell Genesys, Inc. Adenoviral e1a/e1b complementing cell line
US7115391B1 (en) 1999-10-01 2006-10-03 Genovo, Inc. Production of recombinant AAV using adenovirus comprising AAV rep/cap genes
US7132277B1 (en) 2000-01-31 2006-11-07 Merck & Co., Inc. Helper dependent vector system for gene therapy
US7163787B2 (en) 2001-05-07 2007-01-16 Crucell Holland B.V. Methods for the identification of antiviral compounds
US7192759B1 (en) 1999-11-26 2007-03-20 Crucell Holland B.V. Production of vaccines
US7235233B2 (en) 2000-09-26 2007-06-26 Crucell Holland B.V. Serotype 5 adenoviral vectors with chimeric fibers for gene delivery in skeletal muscle cells or myoblasts
WO2007095056A2 (en) 2006-02-09 2007-08-23 Genzyme Corporation Slow intraventricular delivery
US7291484B2 (en) 2003-05-09 2007-11-06 Crucell Holland B.V. Processes for culturing E1-immortalized cells to increase product yields
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
US7300657B2 (en) 2002-12-17 2007-11-27 Crucell Holland B.V. Recombinant viral-based malaria vaccines
WO2008112540A2 (en) * 2007-03-09 2008-09-18 Vectorlogics, Inc. Cells for adenovirus vector and protein production
US7504248B2 (en) 2001-12-07 2009-03-17 Crucell Holland B.V. Production of viruses viral isolates and vaccines
US7521220B2 (en) 1999-11-26 2009-04-21 Crucell Holland B.V. Production of vaccines
US7527961B2 (en) 1999-11-26 2009-05-05 Crucell Holland B.V. Production of vaccines
US7604960B2 (en) 1999-04-15 2009-10-20 Crucell Holland B.V. Transient protein expression methods
EP2163260A2 (en) 2004-01-23 2010-03-17 Istituto di Richerche di Biologia Molecolare P. Angeletti S.p.A. Chimpanzee adenovirus vaccine carriers
EP2168603A1 (en) 2003-11-14 2010-03-31 GenVec, Inc. Therapeutic regimen for treating cancer
US7749493B2 (en) 1998-07-08 2010-07-06 Crucell Holland B.V. Chimeric adenoviruses
WO2010084100A1 (en) 2009-01-20 2010-07-29 Transgene Sa Soluble icam-1 as biomarker for prediction of therapeutic response
WO2010108908A1 (en) 2009-03-24 2010-09-30 Transgene Sa Biomarker for monitoring patients
WO2010119003A1 (en) 2009-04-17 2010-10-21 Transgene Sa Biomarker for monitoring patients
WO2011003615A2 (en) 2009-07-08 2011-01-13 Glycotope Gmbh Perfusion bioreactor
WO2011003905A1 (en) 2009-07-10 2011-01-13 Transgene Sa Biomarker for selecting patients and related methods
WO2011015656A2 (en) 2009-08-07 2011-02-10 Transgene Sa Composition for treating hbv infection
EP2287201A2 (en) 2002-03-01 2011-02-23 Immunomedics, Inc. RS7 antibodies
EP2301968A2 (en) 2002-06-14 2011-03-30 Immunomedics, Inc. Humanized monoclonal antibody HPAM4
WO2011045378A1 (en) * 2009-10-15 2011-04-21 Crucell Holland B.V. Method for the purification of adenovirus particles
WO2011047316A1 (en) 2009-10-16 2011-04-21 The United States Of America, As Represented By The Secretary Department Of Health And Human Services Nucleic acid sequences encoding expandable hiv mosaic proteins
WO2011045381A1 (en) * 2009-10-15 2011-04-21 Crucell Holland B.V. Process for adenovirus purification from high cell density cultures
WO2011057254A2 (en) 2009-11-09 2011-05-12 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Simian adenoviral vector-based vaccines
WO2011057248A2 (en) 2009-11-09 2011-05-12 Genvec, Inc. Simian adenovirus and methods of use
WO2011116189A1 (en) 2010-03-17 2011-09-22 Cornell University Disrupted adenovirus-based vaccine against drugs of abuse
WO2012021730A2 (en) 2010-08-11 2012-02-16 Genvec, Inc. Respiratory syncytial virus (rsv) vaccine
WO2012083297A2 (en) 2010-12-17 2012-06-21 Genvec, Inc. Adenoviral vectors with modified hexon regions
WO2012088041A1 (en) 2010-12-20 2012-06-28 Genvec, Inc. Adenoviral vector-based dengue fever vaccine
US8236561B2 (en) 1999-04-15 2012-08-07 Crucell Holland B.V. Efficient production of IgA in recombinant mammalian cells
WO2012122025A2 (en) 2011-03-04 2012-09-13 Intrexon Corporation Vectors conditionally expressing protein
US8309349B2 (en) 2004-10-28 2012-11-13 University of Pittsburgh—of the Commonwealth System of Higher Education Cell line
US8323663B2 (en) 2005-11-10 2012-12-04 Genvec, Inc. Adenoviral vector-based foot-and-mouth disease vaccine
WO2013007772A1 (en) 2011-07-12 2013-01-17 Transgene Sa Hbv polymerase mutants
EP2567967A2 (en) 2004-04-12 2013-03-13 GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Method of using adenoviral vectors to induce an immune response
WO2013045668A2 (en) 2011-09-29 2013-04-04 Transgene Sa Immunotherapy composition and regimen for treating hepatitis c virus infection
WO2013045658A1 (en) 2011-09-29 2013-04-04 Transgene Sa Immunotherapy composition and regimen for treating hepatitis c virus infection
WO2013052832A2 (en) 2011-10-05 2013-04-11 Genvec, Inc. Adenoviral vectors and methods of use
WO2013052859A2 (en) 2011-10-05 2013-04-11 Genvec, Inc. Adenoviral vector-based respiratory syncytial virus (rsv) vaccine
WO2013052799A2 (en) 2011-10-05 2013-04-11 Genvec, Inc. Adenoviral vectors and methods of use
WO2013052811A2 (en) 2011-10-05 2013-04-11 Genvec, Inc. Adenoviral vectors and methods of use
WO2013116591A1 (en) 2012-02-02 2013-08-08 Genvec, Inc. Adenoviral vector-based malaria vaccine
WO2013119880A1 (en) 2012-02-07 2013-08-15 Global Bio Therapeutics Usa, Inc. Compartmentalized method of nucleic acid delivery and compositions and uses thereof
US8546123B2 (en) 2009-07-16 2013-10-01 Crucell Holland B.V. Production of poliovirus at high titers for vaccine production
WO2013181128A1 (en) 2012-05-29 2013-12-05 Genvec, Inc. Modified serotype 28 adenoviral vectors
WO2013180967A1 (en) 2012-05-29 2013-12-05 Genvec, Inc. Herpes simplex virus vaccine
WO2014009433A1 (en) 2012-07-10 2014-01-16 Transgene Sa Mycobacterium resuscitation promoting factor for use as adjuvant
WO2014009438A2 (en) 2012-07-10 2014-01-16 Transgene Sa Mycobacterial antigen vaccine
WO2014078688A2 (en) 2012-11-16 2014-05-22 Beth Israel Deaconess Medical Center, Inc. Recombinant adenoviruses and use thereof
US8765146B2 (en) 2005-08-31 2014-07-01 Genvec, Inc. Adenoviral vector-based malaria vaccines
ES2525768A1 (en) * 2013-06-24 2014-12-29 Curaxis, S.L. Procedure for the production of glycosylated recombinant proteins. (Machine-translation by Google Translate, not legally binding)
EP2824185A1 (en) 2003-02-24 2015-01-14 GenVec, Inc. Materials and methods for treating disorders of the ear
US8940534B2 (en) 2003-11-03 2015-01-27 Probiogen Ag Immortalized avian cell lines for virus production
WO2015104380A1 (en) 2014-01-09 2015-07-16 Transgene Sa Fusion of heterooligomeric mycobacterial antigens
WO2016037164A1 (en) 2014-09-07 2016-03-10 Selecta Biosciences, Inc. Methods and compositions for attenuating gene expression modulating anti-viral transfer vector immune responses
WO2016131945A1 (en) 2015-02-20 2016-08-25 Transgene Sa Combination product with autophagy modulator
EP3045471B1 (en) 2008-04-16 2017-03-29 Ferring B.V. Recombinant fsh including alpha 2,3- and alpha 2,6-sialylation
WO2017191147A1 (en) 2016-05-04 2017-11-09 Transgene Sa Combination therapy with cpg tlr9 ligand
EP3284821A1 (en) 2010-03-23 2018-02-21 Intrexon Corporation Vectors conditionally expressing therapeutic proteins, host cells comprising the vectors, and uses thereof
WO2018064523A1 (en) 2016-09-30 2018-04-05 Genvec, Inc. Adenovectors for delivery of therapeutic genetic material into t cells
WO2018069316A2 (en) 2016-10-10 2018-04-19 Transgene Sa Immunotherapeutic product and mdsc modulator combination therapy
US9951351B2 (en) 2014-10-09 2018-04-24 Genvec, Inc. Adenoviral vector encoding human atonal homolog-1 (HATH1)
WO2018129268A1 (en) 2017-01-07 2018-07-12 Selecta Biosciences, Inc. Patterned dosing of immunosuppressants coupled to synthetic nanocarriers
US10080783B2 (en) 2006-01-20 2018-09-25 Genzyme Corporation Intraventricular enzyme delivery for lysosomal storage diseases
WO2019075360A1 (en) 2017-10-13 2019-04-18 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector igm responses
WO2020223205A1 (en) 2019-04-28 2020-11-05 Selecta Biosciences, Inc. Methods for treatment of subjects with preexisting immunity to viral transfer vectors
WO2020243261A1 (en) 2019-05-28 2020-12-03 Selecta Biosciences, Inc. Methods and compositions for attenuated anti-viral transfer vector immune response
WO2021215919A1 (en) 2020-04-21 2021-10-28 Polpharma Biologics Utrecht B.V. Humanized anti-human cd89 antibodies and uses thereof
WO2022013221A1 (en) 2020-07-13 2022-01-20 Transgene Treatment of immune depression
US11299713B2 (en) 2017-04-21 2022-04-12 Geneuin-Tech Co., Ltd. Cell line for producing adenovirus and method of preparing the same
WO2022074464A2 (en) 2020-03-05 2022-04-14 Neotx Therapeutics Ltd. Methods and compositions for treating cancer with immune cells
US11364032B2 (en) 2013-08-08 2022-06-21 Global Bio Therapeutics, Inc. Clamp device for minimally invasive procedures and uses thereof
WO2023017494A1 (en) 2021-08-13 2023-02-16 Triovance Holding Llc A skin substitute composition and methods of producing and using the same
US11608362B2 (en) 2018-03-06 2023-03-21 Precigen, Inc. Hepatitis B vaccines and uses of the same
WO2023064367A1 (en) 2021-10-12 2023-04-20 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector igm responses
WO2023172624A1 (en) 2022-03-09 2023-09-14 Selecta Biosciences, Inc. Immunosuppressants in combination with anti-igm agents and related dosing
US11773142B2 (en) 2017-12-11 2023-10-03 Beth Israel Deaconess Medical Center, Inc. Recombinant adenoviruses and uses thereof
WO2023213764A1 (en) 2022-05-02 2023-11-09 Transgene Fusion polypeptide comprising an anti-pd-l1 sdab and a member of the tnfsf

Families Citing this family (232)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0872552A1 (en) * 1997-04-15 1998-10-21 Leadd B.V. A gene delivery vehicle expressing the apoptosis-inducing proteins VP2 and/or Apoptin
FR2705686B1 (en) * 1993-05-28 1995-08-18 Transgene Sa New defective adenoviruses and corresponding complementation lines.
US6080569A (en) * 1993-06-24 2000-06-27 Merck & Co., Inc. Adenovirus vectors generated from helper viruses and helper-dependent vectors
DE69533295T3 (en) * 1994-02-16 2009-07-16 The Government Of The United States Of America, As Represented By The Secretary, The Department Of Health And Human Services Melanoma-associated antigens, epitopes thereof and melanoma-containing vaccines
US7252989B1 (en) * 1994-04-04 2007-08-07 Board Of Regents, The University Of Texas System Adenovirus supervector system
US5998205A (en) * 1994-11-28 1999-12-07 Genetic Therapy, Inc. Vectors for tissue-specific replication
US6281010B1 (en) * 1995-06-05 2001-08-28 The Trustees Of The University Of Pennsylvania Adenovirus gene therapy vehicle and cell line
US6783980B2 (en) * 1995-06-15 2004-08-31 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
US20020068049A1 (en) * 1998-09-10 2002-06-06 Henderson Daniel R. Tissue specific adenoviral vectors
US20040156861A1 (en) * 1996-07-11 2004-08-12 Figdor Carl Gustav Melanoma associated peptide analogues and vaccines against melanoma
WO1998021350A1 (en) * 1996-11-13 1998-05-22 Board Of Regents, The University Of Texas System Diminishing viral gene expression by promoter replacement
US6403370B1 (en) 1997-02-10 2002-06-11 Genstar Therapeutics Corporation Oncolytic/immunogenic complementary-adenoviral vector system
US6696423B1 (en) 1997-08-29 2004-02-24 Biogen, Inc. Methods and compositions for therapies using genes encoding secreted proteins such as interferon-beta
US20040043489A1 (en) * 1998-07-08 2004-03-04 Menzo Havenga Gene delivery vectors provided with a tissue tropism for dendritic cells and methods of use
US6900049B2 (en) * 1998-09-10 2005-05-31 Cell Genesys, Inc. Adenovirus vectors containing cell status-specific response elements and methods of use thereof
US20050164386A1 (en) * 1999-04-15 2005-07-28 Uytdehaag Alphonsus G. Overexpression of enzymes involved in post-translational protein modifications in human cells
US20060099685A1 (en) * 1999-04-15 2006-05-11 Yallop Christopher A Recombinant expression of factor VIII in human cells
US20050170463A1 (en) * 1999-04-15 2005-08-04 Abraham Bout Recombinant protein production in permanent amniocytic cells that comprise nucleic acid encoding adenovirus E1A and E1B proteins
ATE364707T1 (en) 1999-05-17 2007-07-15 Crucell Holland Bv RECOMBINANT HUMAN ADENOVIRUS SEROTYPE 35
US20050232900A1 (en) * 1999-05-18 2005-10-20 Crucell Holland B.V. Serotype of adenovirus and uses thereof
EP1067190A1 (en) * 1999-07-09 2001-01-10 Introgene B.V. Gene therapy for enhancing and/or inducing angiogenesis
AU1817401A (en) * 1999-12-14 2001-06-25 Genovo, Incorporated Methods and compositions for the manufacture of replication incompetent adenovirus
CA2396779A1 (en) * 2000-01-07 2001-07-12 Stichting Klinishce Farmacologie Groningen Gene therapy to promote angiogenesis and/or the treatment of heart failure
AU2001245944B2 (en) 2000-03-24 2007-01-18 Cell Genesys, Inc. Human urothelial cell specific uroplakin transcriptional regulatory sequences, vectors comprising the same, and methods of use thereof
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
EP1157999A1 (en) * 2000-05-24 2001-11-28 Introgene B.V. Methods and means for enhancing skin transplantation using gene delivery vehicles having tropism for primary fibroblasts, as well as other uses thereof
KR20010064682A (en) * 2000-07-01 2001-07-11 김재호 Double Suicide Gene System using E1B attenuated adenovirus
US20020164333A1 (en) * 2000-07-10 2002-11-07 The Scripps Research Institute Bifunctional molecules and vectors complexed therewith for targeted gene delivery
US7223406B2 (en) * 2000-07-21 2007-05-29 The Regents Of The University Of California Methods and compositions for preventing and treating male erectile dysfunction and female sexual arousal disorder
EP1365794A2 (en) 2000-07-21 2003-12-03 Lue, Tom Prevention and treatment of sexual arousal disorders
US20080194022A1 (en) * 2000-08-03 2008-08-14 Clarke Michael F Isolation and use of solid tumor stem cells
US8044259B2 (en) 2000-08-03 2011-10-25 The Regents Of The University Of Michigan Determining the capability of a test compound to affect solid tumor stem cells
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
US20040101957A1 (en) * 2001-09-14 2004-05-27 Emini Emilio A. Enhanced first generation adenovirus vaccines expressing codon optimized hiv1-gag, pol.nef and modifications
NZ524743A (en) * 2000-09-20 2004-07-30 Crucell Holland B Gene delivery vectors provided with a tissue tropism for dendritic cells provided by a viral capsid protein
CA2439185A1 (en) * 2001-02-23 2002-09-06 Novartis Ag Vector constructs
JP2004533815A (en) 2001-03-14 2004-11-11 ミリアド・ジェネティックス・インコーポレイテッド TSG101-GAG interaction and use thereof
US7229774B2 (en) 2001-08-02 2007-06-12 Regents Of The University Of Michigan Expression profile of prostate cancer
US20030119771A1 (en) * 2001-08-22 2003-06-26 Rompaey Luc Van Modulators of bone homeostasis identified in a high-throughput screen
DK1436397T3 (en) * 2001-10-11 2010-08-09 Merck Sharp & Dohme Hepatitis C virus vaccine
ES2344253T3 (en) 2001-10-11 2010-08-23 MERCK SHARP & DOHME CORP. VACCINE AGAINST THE VIRUS OF HEPATITIS C.
JP4583029B2 (en) 2001-10-29 2010-11-17 クルセル ホランド ベー ヴェー Method and method for producing a protein having a predetermined post-translational modification
DK1456238T3 (en) * 2001-12-17 2008-08-18 Crucell Holland Bv Production of F (ab ') 2 fragments in mammalian cells
US20030165954A1 (en) 2002-01-09 2003-09-04 Third Wave Technologies, Inc. Cancer profiles
JP4488290B2 (en) * 2002-01-24 2010-06-23 ザ・スクリップス・リサーチ・インスティテュート Fiber shaft mutation for efficient targeting
EP1474533B1 (en) * 2002-02-20 2006-04-26 Merck & Co., Inc. Method of determining adenovirus particle concentration
CA2478901A1 (en) * 2002-03-29 2003-10-16 Merck & Co., Inc. Methods of virus production
WO2003087348A1 (en) * 2002-04-08 2003-10-23 The Johns Hopkins University Packaging cell line for diphtheria toxin expressing non-replicating adenovirus
EA010828B1 (en) * 2002-04-25 2008-12-30 Круселл Холланд Б.В. Recombinant adenovirus vectors and methods for the production thereof and use
IL164802A0 (en) * 2002-04-25 2005-12-18 Crucell Holland Bv Stable adenoviral vectors and methods for propagation thereof
US20030219696A1 (en) * 2002-05-23 2003-11-27 Moreland Gerald W. Method and apparatus for preventing backflow in dental saliva evacuators
US7226755B1 (en) * 2002-09-25 2007-06-05 The Procter & Gamble Company HPTPbeta as a target in treatment of angiogenesis mediated disorders
US7507568B2 (en) * 2002-09-25 2009-03-24 The Proctor & Gamble Company Three dimensional coordinates of HPTPbeta
US20080153083A1 (en) * 2003-10-23 2008-06-26 Crucell Holland B.V. Settings for recombinant adenoviral-based vaccines
AU2003288273A1 (en) 2002-10-23 2004-05-13 Crucell Holland B.V. New settings for recombinant adenoviral-based vaccines
US20050221493A1 (en) * 2002-12-04 2005-10-06 Crucell Holland B.V. Recombinant virus production for the manufacturing of vaccines
ATE354645T1 (en) * 2002-12-20 2007-03-15 Chromagenics Bv MEANS AND METHODS FOR PRODUCING A PROTEIN THROUGH CHROMATIN OPENERS WHICH CAN MAKE CHROMATIN MORE ACCESSIBLE TO TRANSCRIPTION FACTORS
EP1594459B1 (en) 2002-12-30 2010-02-17 Angiotech International Ag Drug delivery from rapid gelling polymer composition
US20070212675A1 (en) * 2003-04-15 2007-09-13 Novartis Ag Flap Endonuclease (Fen1) Regulatory Sequences And Uses Thereof
WO2004092397A2 (en) * 2003-04-15 2004-10-28 Novartis Ag Tmprss2 regulatory sequences and uses thereof
DK1626992T3 (en) * 2003-05-23 2010-09-20 Crucell Holland Bv Production of recombinant IgM in PER.C6 cells
US7709224B2 (en) * 2003-06-03 2010-05-04 Biosante Pharmaceuticals, Inc. Compositions and methods for enhanced expression of recombinant polypeptides from a single vector using a peptide cleavage site
US20050136035A1 (en) * 2003-06-03 2005-06-23 Derek Ko Cell specific replication-competent viral vectors comprising a self processing peptide cleavage site
US20060019256A1 (en) * 2003-06-09 2006-01-26 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US7261882B2 (en) 2003-06-23 2007-08-28 Reagents Of The University Of Colorado Methods for treating neuropathic pain by administering IL-10 polypeptides
US20050186178A1 (en) * 2003-08-28 2005-08-25 Ennist David L. Oncolytic adenoviral vectors encoding GM-CSF
CN1863918B (en) * 2003-10-02 2011-03-30 克鲁塞尔荷兰公司 Packaging cells for recombinant adenovirus
US7482156B2 (en) * 2003-10-15 2009-01-27 Cell Genesys, Inc. Hepatocellular carcinoma specific promoter and uses thereof
US20050232927A1 (en) * 2004-02-03 2005-10-20 The Regents Of The University Of Michigan Compositions and methods for characterizing, regulating, diagnosing, and treating cancer
JP2007522814A (en) * 2004-02-23 2007-08-16 クルセル ホランド ベー ヴェー Virus purification method
US7473418B2 (en) * 2004-03-25 2009-01-06 Cell Genesys, Inc. Pan cancer oncolytic vectors and methods of use thereof
WO2005115477A2 (en) * 2004-04-13 2005-12-08 Quintessence Biosciences, Inc. Non-natural ribonuclease conjugates as cytotoxic agents
EP1773303A2 (en) * 2004-05-25 2007-04-18 Chimeracore, Inc. Self-assembling nanoparticle drug delivery system
US7858323B2 (en) 2004-06-09 2010-12-28 The Regents Of The University Of Michigan Phage microarray profiling of the humoral response to disease
US20060062764A1 (en) * 2004-08-25 2006-03-23 Seshidar-Reddy Police Fiber-modified adenoviral vectors for enhanced transduction of tumor cells
EP1784493A2 (en) * 2004-09-01 2007-05-16 The Government of the United States of America as Represented by The Department of Health and Human Services Adenoviral vectors able to transduce apcs, potential use in immune response generation
PT1807009E (en) 2004-10-05 2015-02-25 Univ California Stepped cannula
US8563511B2 (en) 2004-10-06 2013-10-22 University Of Rochester Treatment of pulmonary hypertension using an agent that inhibits a tissue factor pathway
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
MX2007004595A (en) * 2004-11-08 2007-08-15 Chromagenics Bv 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
US20060195935A1 (en) 2004-11-08 2006-08-31 Chromagenics B.V. Selection of host cells expressing protein at high levels
CA2590943C (en) * 2004-12-13 2013-10-22 Canji, Inc. Cell lines for production of replication-defective adenovirus
EP1856526A4 (en) 2005-01-20 2008-11-12 Univ Rochester Thioredoxin interacting protein (txnip) as regulator of vascular function
AU2006212885A1 (en) * 2005-02-11 2006-08-17 Merck & Co., Inc. Adenovirus serotype 26 vectors, nucleic acid and viruses produced thereby
ES2317517T5 (en) * 2005-04-11 2016-01-21 Crucell Holland B.V. Virus purification using ultrafiltration
US20060234347A1 (en) * 2005-04-13 2006-10-19 Harding Thomas C Targeting multiple angiogenic pathways for cancer therapy using soluble tyrosine kinase receptors
US8524678B2 (en) 2005-05-31 2013-09-03 The Regents Of The University Of Colorado, A Body Corporate Method for delivering genes
AU2006259583A1 (en) * 2005-06-13 2006-12-28 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
WO2006135886A2 (en) * 2005-06-13 2006-12-21 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
CA2610919C (en) 2005-06-17 2013-09-10 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Hepatitis c virus nucleic acid vaccine
AU2006271666B2 (en) * 2005-07-22 2011-04-07 Crucell Holland B.V. Cell line for producing coronaviruses
US9651543B2 (en) 2005-08-31 2017-05-16 The United States Of America As Represented By The Secretary Of The Navy Malaria antigen screening method
US8450055B2 (en) * 2005-08-31 2013-05-28 The United States Of America As Represented By The Secretary Of The Navy Malaria antigen screening method
US7718369B2 (en) 2005-09-12 2010-05-18 The Regents Of The University Of Michigan Recurrent gene fusions in prostate cancer
WO2007047907A2 (en) * 2005-10-14 2007-04-26 The Regents Of The University Of Michigan Dek protein compositions and methods of using the same
US7794951B2 (en) * 2005-10-18 2010-09-14 University Of Massachusetts Medical School SREBP2gc transcription factors and uses thereof
US7723477B2 (en) 2005-10-31 2010-05-25 Oncomed Pharmaceuticals, Inc. Compositions and methods for inhibiting Wnt-dependent solid tumor cell growth
AU2006308847C1 (en) * 2005-10-31 2012-05-10 Oncomed Pharmaceuticals, Inc. Compositions and methods for treating and diagnosing cancer
JP2009513708A (en) 2005-10-31 2009-04-02 オンコメッド ファーマシューティカルズ インコーポレイテッド Compositions and methods for diagnosis and treatment of cancer
EP1974044B1 (en) * 2005-12-12 2011-01-26 CANJI, Inc. Adenoviral expression vectors having an expression cassette in the e1 region and an inactivated e2b polymerase
WO2008121102A2 (en) * 2006-02-21 2008-10-09 The Regents Of The University Of Michigan Hedgehog signaling pathway antagonist cancer treatment
US7968700B2 (en) * 2006-03-20 2011-06-28 Chromagenics B.V. Expression augmenting DNA fragments, use thereof, and methods for finding thereof
DE602007005366D1 (en) * 2006-04-07 2010-04-29 Chimeros Inc COMPOSITIONS AND METHOD FOR THE TREATMENT OF B-CELL MALIGNOMES
ES2643469T3 (en) 2006-04-07 2017-11-23 Aerpio Therapeutics, Inc. Antibodies that bind to the human tyrosine phosphatase beta protein (HPTP-beta) and uses thereof
EP2377953A1 (en) 2006-06-23 2011-10-19 Myriad Genetics, Inc. DPYD gene variants and use thereof
WO2007149594A2 (en) * 2006-06-23 2007-12-27 Quintessence Biosciences, Inc. Modified ribonucleases
US7795444B2 (en) * 2006-06-27 2010-09-14 Warner Chilcott Company Human protein tyrosine phosphatase inhibitors and methods of use
US8846685B2 (en) 2006-06-27 2014-09-30 Aerpio Therapeutics Inc. Human protein tyrosine phosphatase inhibitors and methods of use
US7622593B2 (en) 2006-06-27 2009-11-24 The Procter & Gamble Company Human protein tyrosine phosphatase inhibitors and methods of use
US7589212B2 (en) * 2006-06-27 2009-09-15 Procter & Gamble Company Human protein tyrosine phosphatase inhibitors and methods of use
SI2634242T1 (en) 2006-07-14 2017-07-31 Patheon Holdings I B.V. Improved process for the culturing of cells
JP2009543868A (en) 2006-07-17 2009-12-10 クインテセンス バイオサイエンシーズ インコーポレーティッド Methods and compositions for cancer treatment
WO2008030605A2 (en) * 2006-09-08 2008-03-13 The Regents Of The University Of Michigan Herv group ii viruses in lymphoma and cancer
EP2114445A2 (en) * 2007-01-09 2009-11-11 Genvec, Inc. Adenoviral vector-based malaria vaccines
US11371194B2 (en) * 2007-01-19 2022-06-28 Brock Usa, Llc Base for turf system
WO2008092002A2 (en) 2007-01-24 2008-07-31 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing pancreatic cancer
CA2683063A1 (en) * 2007-04-09 2008-10-16 Chimeros, Inc. Self-assembling nanoparticle drug delivery system
US10745701B2 (en) 2007-06-28 2020-08-18 The Trustees Of Princeton University Methods of identifying and treating poor-prognosis cancers
US20090324596A1 (en) 2008-06-30 2009-12-31 The Trustees Of Princeton University Methods of identifying and treating poor-prognosis cancers
WO2009009432A2 (en) 2007-07-06 2009-01-15 The Regents Of The University Of Michigan Recurrent gene fusions in prostate cancer
WO2009026152A1 (en) 2007-08-16 2009-02-26 The Regents Of The University Of Michigan Metabolomic profiling of prostate cancer
DE102007041655A1 (en) 2007-09-03 2009-03-05 Medicyte Gmbh Propagation of primary cells and their use
CA2702043A1 (en) * 2007-10-08 2009-04-16 Quintessence Biosciences, Inc. Compositions and methods for ribonuclease-based therapies
US8518884B2 (en) 2008-04-25 2013-08-27 Northwestern University Methods for treating atrial or ventricular arrhythmias by administering a G-protein alpha inhibitor
US8193151B2 (en) * 2008-04-25 2012-06-05 Northwestern University Methods for treating atrial or ventricular arrhythmias
KR101639454B1 (en) * 2008-07-15 2016-07-13 크루셀 홀란드 비.브이. Scalable process for culturing per.c6 cells and producing products therefrom
EP2342321B1 (en) 2008-09-17 2018-04-11 Isogenis, Inc. Construction of fully-deleted adenovirus-based gene delivery vectors and uses thereof
BRPI0919473A2 (en) 2008-09-26 2017-08-29 Oncomed Pharm Inc FRIZZLED BINDING AGENTS AND THEIR USES
WO2010039985A1 (en) 2008-10-01 2010-04-08 Quintessence Biosciences, Inc. Therapeutic Ribonucleases
US20110184154A1 (en) * 2008-10-17 2011-07-28 Percivia Llc Cell broth clarification and host cell protein removal
CN102203242B (en) * 2008-11-03 2013-06-12 克鲁塞尔荷兰公司 Method for the production of adenoviral vectors
CA2749113A1 (en) 2009-01-09 2010-07-15 The Regents Of The University Of Michigan Recurrent gene fusions in cancer
US9096555B2 (en) 2009-01-12 2015-08-04 Aerpio Therapeutics, Inc. Methods for treating vascular leak syndrome
WO2010107991A2 (en) 2009-03-18 2010-09-23 Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services Use of ixolaris, a tissue factor inhibitor, for the treatment and prevention of cancer
WO2010120874A2 (en) 2009-04-14 2010-10-21 Chimeros, Inc. Chimeric therapeutics, compositions, and methods for using same
CA2756591C (en) 2009-04-23 2018-11-06 Crucell Holland B.V. Recombinant human alpha1-antitrypsin
US8748386B2 (en) 2009-06-10 2014-06-10 New York University Immunological targeting of pathological Tau proteins
CA2748765C (en) 2009-07-06 2014-07-22 Akebia Therapeutics Inc. Compounds, compositions, and methods for preventing metastasis of cancer cells
US8883832B2 (en) 2009-07-06 2014-11-11 Aerpio Therapeutics Inc. Compounds, compositions, and methods for preventing metastasis of cancer cells
US9605844B2 (en) * 2009-09-01 2017-03-28 Cree, Inc. Lighting device with heat dissipation elements
EP2521566B1 (en) 2010-01-08 2014-09-03 ProFibrix BV Fibrinogen preparations enriched in fibrinogen with an extended alpha chain
TWI535445B (en) 2010-01-12 2016-06-01 安可美德藥物股份有限公司 Wnt antagonists and methods of treatment and screening
CN102762721B (en) 2010-02-15 2015-03-11 克鲁塞尔荷兰公司 Method for the production of Ad26 adenoviral vectors
EP2548025A4 (en) * 2010-03-17 2013-09-25 Univ Michigan Using phage epitopes to profile the immune response
CA2794674A1 (en) 2010-04-01 2011-10-06 Oncomed Pharmaceuticals, Inc. Frizzled-binding agents and uses thereof
WO2012030512A1 (en) 2010-09-03 2012-03-08 Percivia Llc. Flow-through protein purification process
EP2618838A1 (en) 2010-09-20 2013-07-31 Crucell Holland B.V. Therapeutic vaccination against active tuberculosis
US9168292B2 (en) 2010-09-27 2015-10-27 Crucell Holland B.V. Heterologous prime boost vaccination regimen against malaria
DE102010041958A1 (en) 2010-10-04 2012-04-05 Medicyte Gmbh Suitable hepatocytes for in vitro genotoxicity tests
US20130344053A1 (en) 2010-12-28 2013-12-26 University Of Rochester Methods of Modifying Insulin Signaling Using Biliverdin Reductase (BVR) and BVR Derived Peptides
WO2012106281A2 (en) 2011-01-31 2012-08-09 The General Hospital Corporation Multimodal trail molecules and uses in cellular therapies
WO2013001372A2 (en) 2011-06-30 2013-01-03 University Of Oslo Methods and compositions for inhibition of activation of regulatory t cells
HUE030501T2 (en) 2011-07-06 2017-05-29 Sykehuset Sorlandet Hf Egfr targeted therapy
EP3741384A1 (en) 2011-09-07 2020-11-25 Mount Sinai School Of Medicine Ceramidase and cell differentiation
AU2012305714A1 (en) 2011-09-09 2014-03-27 Biomed Realty, L.P. Methods and compositions for controlling assembly of viral proteins
EP2780034A1 (en) 2011-11-14 2014-09-24 Crucell Holland B.V. Heterologous prime-boost immunization using measles virus-based vaccines
JP5969044B2 (en) 2011-11-24 2016-08-10 バイロメッド カンパニー リミテッド Adenovirus-producing new cell line and its use
US20150004144A1 (en) 2011-12-02 2015-01-01 The General Hospital Corporation Differentiation into brown adipocytes
CN104136613B (en) 2011-12-08 2017-12-29 威洛克有限公司 Carrier, method with virulent gene and application thereof
US8932607B2 (en) 2012-03-12 2015-01-13 Crucell Holland B.V. Batches of recombinant adenovirus with altered terminal ends
ES2582504T3 (en) 2012-03-12 2016-09-13 Crucell Holland B.V. Lots of recombinant adenovirus with altered ends
KR102050616B1 (en) 2012-03-22 2019-12-03 얀센 백신스 앤드 프리벤션 비.브이. Vaccine against rsv
US9125870B2 (en) 2012-03-22 2015-09-08 Crucell Holland B.V. Vaccine against RSV
US9982240B2 (en) 2012-08-03 2018-05-29 Sanofi Pasteur Production of infectious influenza viruses
EP2890720B1 (en) 2012-08-30 2019-07-17 The General Hospital Corporation Compositions and methods for treating cancer
US9694050B2 (en) 2012-10-21 2017-07-04 University Of Rochester THY1 (CD90) as a novel therapy to control adipose tissue accumulation
JP2015536933A (en) 2012-10-23 2015-12-24 オンコメッド ファーマシューティカルズ インコーポレイテッド Methods of treating neuroendocrine tumors using Wnt pathway binding agents
KR102228358B1 (en) 2012-12-21 2021-03-16 쉬케후세트 솔란데트 에이치에프 Egfr targeted therapy of neurological disorders and pain
CN105073195A (en) 2013-02-04 2015-11-18 昂科梅德制药有限公司 Methods and monitoring of treatment with a Wnt pathway inhibitor
US9168300B2 (en) 2013-03-14 2015-10-27 Oncomed Pharmaceuticals, Inc. MET-binding agents and uses thereof
KR101606910B1 (en) 2013-03-29 2016-04-04 충북보건과학대학교 산학협력단 A549 Based Cell Line for Preparing Adenovirus Vector
SG10201912901YA (en) 2013-04-17 2020-02-27 Genzyme Corp Compositions and methods for treating and preventing macular degeneration
WO2014176259A1 (en) 2013-04-22 2014-10-30 Icahn School Of Medicine At Mount Sinai Mutations in pdgfrb and notch3 as causes of autosomal dominant infantile myofibromatosis
MY181066A (en) 2013-04-25 2020-12-17 Janssen Vaccines & Prevention Bv Stabilized soluble pre-fusion rsv f polypeptides
EP3848044A1 (en) 2013-05-21 2021-07-14 President and Fellows of Harvard College Engineered heme-binding compositions and uses thereof
KR102313153B1 (en) 2013-06-17 2021-10-15 얀센 백신스 앤드 프리벤션 비.브이. Stabilized soluble pre-fusion rsv f polypeptides
AP2016009154A0 (en) 2013-09-19 2016-04-30 Crucell Holland Bv Improved adenovirus formulations
SG10201808061WA (en) 2013-09-30 2018-10-30 Janssen Vaccines & Prevention Bv Method for the clarification of high density crude cell culture harvest
TWI687225B (en) 2014-02-06 2020-03-11 美商健臻公司 Compositions and methods for treating and preventing macular degeneration
EP2927685A1 (en) 2014-04-02 2015-10-07 Medicyte GmbH Suitable hepatocytes for in-vitro hepatitis tests
US10294460B2 (en) 2014-07-24 2019-05-21 Janssen Vaccines & Prevention B.V. Process for the purification of poliovirus from cell cultures
WO2016044707A1 (en) 2014-09-18 2016-03-24 Cedars-Sinai Medical Center Compositions and methods for treating fibrosis
CN107075521B (en) 2014-11-04 2021-06-08 扬森疫苗与预防公司 Therapeutic HPV16 vaccine
EP3259374B1 (en) 2015-02-19 2019-08-21 Janssen Vaccines & Prevention B.V. Method for quantification of virus particles using capillary zone electrophoresis.
EP3271729B1 (en) 2015-03-18 2020-09-23 Janssen Vaccines & Prevention B.V. Assays for recombinant expression systems
SI3283634T1 (en) 2015-04-14 2019-08-30 Janssen Vaccines & Prevention B.V. Recombinant adenovirus expressing two transgenes with a bidirectional promoter
US20180112006A1 (en) 2015-04-17 2018-04-26 The General Hospital Corporation Agents, systems and methods for treating cancer
CN114366802A (en) 2015-04-22 2022-04-19 西达-赛奈医疗中心 Enterally delivered bitter oligopeptides for the treatment of type 2 diabetes
KR20180026734A (en) 2015-07-07 2018-03-13 얀센 백신스 앤드 프리벤션 비.브이. Stabilized soluble fusion-pre-RSV F polypeptide
EA039065B1 (en) 2015-07-07 2021-11-29 Янссен Вэксинс Энд Превеншн Б.В. Vaccine against rsv
EP3337500A1 (en) 2015-08-20 2018-06-27 Janssen Vaccines & Prevention B.V. Therapeutic hpv18 vaccines
EA036412B1 (en) 2015-10-06 2020-11-09 Янссен Вэксинс Энд Превеншн Б.В. Method for protecting an adenovirus from plastic-induced degradation and use of 2-hydroxypropyl--cyclodextrin for protecting the adenovirus
WO2017074517A1 (en) * 2015-10-30 2017-05-04 Seracare Life Sciences, Inc. Adenovirus control virus
US10377801B2 (en) 2015-11-04 2019-08-13 Northwestern University Amelioration of chronic kidney disease
US11273151B2 (en) 2015-11-04 2022-03-15 Icahn School Of Medicine At Mount Sinai Methods of treating tumors and cancer, and identifying candidate subjects for such treatment
SG11201807913XA (en) 2016-04-05 2018-10-30 Janssen Vaccines & Prevention Bv Stabilized soluble pre-fusion rsv f proteins
MY193584A (en) 2016-04-05 2022-10-19 Janssen Vaccines & Prevention Bv Vaccine against rsv
EP3448881B1 (en) 2016-04-26 2023-06-07 Icahn School of Medicine at Mount Sinai Treatment of hippo pathway mutant tumors and methods of identifying subjects as candidates for treatment
JP7053491B2 (en) 2016-05-02 2022-04-12 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 Therapeutic HPV vaccine combination
KR102573534B1 (en) 2016-05-12 2023-08-31 얀센 백신스 앤드 프리벤션 비.브이. Strong and balanced bidirectional promoter
ES2836598T3 (en) 2016-05-30 2021-06-25 Janssen Vaccines & Prevention Bv Stabilized prefusion RSV F proteins
US11001858B2 (en) 2016-06-20 2021-05-11 Janssen Vaccines & Prevention B.V. Potent and balanced bidirectional promoter
WO2018013509A1 (en) 2016-07-11 2018-01-18 Arizona Board Of Regents On Behalf Of The University Of Arizona Compositions and methods for diagnosing and treating arrhythmias
US10744196B2 (en) 2016-07-14 2020-08-18 Janssen Vaccines & Prevention B.V. HPV vaccines
WO2018140630A1 (en) 2017-01-25 2018-08-02 Northwestern University Autophagy inducers for treatment of cns conditions
CA3053212C (en) 2017-02-09 2021-04-13 Janssen Vaccines & Prevention B.V. Potent and short promoter for expression of heterologous genes
CN110914413A (en) 2017-02-17 2020-03-24 隆萨有限公司 Mammalian cell producing adeno-associated virus
JP2020519663A (en) 2017-05-17 2020-07-02 ヤンセン ファッシンズ アンド プリベンション ベーフェーJanssen Vaccines & Prevention B.V. Methods and compositions for inducing protective immunity against RSV infection
MX2020002876A (en) 2017-09-15 2020-07-22 Janssen Vaccines & Prevention Bv Method for the safe induction of immunity against rsv.
MX2020004490A (en) 2017-10-31 2020-08-13 Janssen Vaccines & Prevention Bv Adenovirus vectors and uses thereof.
MX2020004487A (en) 2017-10-31 2020-08-13 Janssen Vaccines & Prevention Bv Adenovirus and uses thereof.
SG11202003200VA (en) 2017-10-31 2020-05-28 Janssen Vaccines & Prevention Bv Adenovirus and uses thereof
KR20200074987A (en) 2017-10-31 2020-06-25 얀센 백신스 앤드 프리벤션 비.브이. Adenovirus and uses thereof
AU2018372906A1 (en) 2017-11-22 2020-06-11 The Regents Of The University Of Michigan Compositions and methods for treating cancer
WO2019145310A1 (en) 2018-01-23 2019-08-01 Janssen Vaccines & Prevention B.V. Influenza virus vaccines 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
US20220204567A1 (en) 2019-04-25 2022-06-30 Janssen Vaccines & Prevention B.V. Recombinant influenza antigens
KR20220008875A (en) 2019-05-15 2022-01-21 얀센 백신스 앤드 프리벤션 비.브이. Co-administration of seasonal influenza vaccine and adenovirus-based respiratory syncytial virus vaccine
EA202193119A1 (en) 2019-05-15 2022-02-18 Янссен Вэксинс Энд Превеншн Б.В. PREVENTIVE TREATMENT OF RESPIRATORY SYNCYTHIAL VIRUS INFECTION WITH ADENOVIRUS VACCINE
EP3969046A1 (en) 2019-05-16 2022-03-23 Janssen Vaccines & Prevention B.V. Methods for inducing a safe immune response against polio virus
EP4025247A1 (en) 2019-09-05 2022-07-13 Janssen Vaccines & Prevention B.V. Influenza virus vaccines and uses thereof
CN114710957A (en) 2019-10-03 2022-07-05 扬森疫苗与预防公司 Adenovirus vectors and uses thereof
MX2022005980A (en) 2019-11-18 2022-09-07 Janssen Biotech Inc Vaccines based on mutant calr and jak2 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
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
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
WO2022009049A1 (en) 2020-07-06 2022-01-13 Janssen Biotech, Inc. Prostate neoantigens and their uses
US20230029453A1 (en) 2020-07-06 2023-02-02 Janssen Biotech, Inc. Prostate Neoantigens And Their Uses
WO2022009051A1 (en) 2020-07-06 2022-01-13 Janssen Biotech, Inc. A method for determining responsiveness to prostate cancer treatment
EP4196137A1 (en) 2020-08-11 2023-06-21 Musculoskeletal Transplant Foundation Method for treating cardiac conditions with placenta-derived compositions

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994028152A1 (en) * 1993-05-28 1994-12-08 Transgene S.A. Defective adenoviruses and corresponding complementation lines
WO1995002697A1 (en) * 1993-07-13 1995-01-26 Rhone-Poulenc Rorer S.A. Defective adenovirus vectors and use thereof in gene therapy
WO1995027071A2 (en) * 1994-04-04 1995-10-12 Board Of Regents, The University Of Texas System An adenovirus supervector system
WO1995034671A1 (en) * 1994-06-10 1995-12-21 Genvec, Inc. Complementary adenoviral vector systems and cell lines

Family Cites Families (162)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US545522A (en) * 1895-09-03 Vehicle-wheel
US4497796A (en) 1980-03-26 1985-02-05 The Regents Of The University Of California Gene transfer in intact mammals
US4727028A (en) 1981-06-22 1988-02-23 Eli Lilly And Company Recombinant DNA cloning vectors and the eukaryotic and prokaryotic transformants thereof
US4405712A (en) 1981-07-01 1983-09-20 The United States Of America As Represented By The Department Of Health And Human Services LTR-Vectors
US4593002A (en) * 1982-01-11 1986-06-03 Salk Institute Biotechnology/Industrial Associates, Inc. Viruses with recombinant surface proteins
US4487829A (en) * 1982-03-23 1984-12-11 Massachusetts Institute Of Technology Production and use of monoclonal antibodies against adenoviruses
US4792525A (en) * 1982-08-04 1988-12-20 La Jolla Cancer Research Foundation Tetrapeptide
US4517686A (en) * 1982-08-04 1985-05-21 La Jolla Cancer Research Foundation Polypeptide
US4589881A (en) * 1982-08-04 1986-05-20 La Jolla Cancer Research Foundation Polypeptide
US4578079A (en) * 1982-08-04 1986-03-25 La Jolla Cancer Research Foundation Tetrapeptide
US5190931A (en) 1983-10-20 1993-03-02 The Research Foundation Of State University Of New York Regulation of gene expression by employing translational inhibition of MRNA utilizing interfering complementary MRNA
US5208149A (en) 1983-10-20 1993-05-04 The Research Foundation Of State University Of New York Nucleic acid constructs containing stable stem and loop structures
US4703008A (en) 1983-12-13 1987-10-27 Kiren-Amgen, Inc. DNA sequences encoding erythropoietin
NZ210501A (en) 1983-12-13 1991-08-27 Kirin Amgen Inc Erythropoietin produced by procaryotic or eucaryotic expression of an exogenous dna sequence
US4740463A (en) 1984-04-13 1988-04-26 Massachusetts Institute Of Technology Methods and artificial genes for antagonizing the function of an oncogene
FR2573436B1 (en) 1984-11-20 1989-02-17 Pasteur Institut RECOMBINANT DNA COMPRISING A NUCLEOTIDE SEQUENCE ENCODING A DETERMINED POLYPEPTIDE UNDER THE CONTROL OF AN ADENOVIRUS PROMOTER, VECTORS CONTAINING THIS RECOMBINANT DNA, EUKARYOT CELLS TRANSFORMED BY THIS RECOMBINANT DNA, THE CONSTITUTION OF VACCINES
IL77081A (en) 1984-12-04 1999-10-28 Genetics Inst Dna sequence encoding human erythropoietin process for the preparation thereof and a pharmaceutical composition of human erythropoietin
US4797368A (en) * 1985-03-15 1989-01-10 The United States Of America As Represented By The Department Of Health And Human Services Adeno-associated virus as eukaryotic expression vector
US5744133A (en) 1986-08-13 1998-04-28 Transgene S.A. Expression of a tumor-specific antigen by a recombinant vector virus and use thereof in preventitive or curative treatment of the corresponding tumor
US6007806A (en) 1986-08-13 1999-12-28 Transgene S.A. Expression of a tumor-specific antigen by a recombinant vector virus and use thereof in preventive or curative treatment of the corresponding tumor
US4835260A (en) 1987-03-20 1989-05-30 Genetics Institute, Inc. Erythropoietin composition
US5166320A (en) * 1987-04-22 1992-11-24 University Of Connecticut Carrier system and method for the introduction of genes into mammalian cells
US4956281A (en) * 1987-06-03 1990-09-11 Biogen, Inc. DNA sequences, recombinant DNA molecules and processes for producing lymphocyte function associated antigen-3
US5024939A (en) * 1987-07-09 1991-06-18 Genentech, Inc. Transient expression system for producing recombinant protein
US5378618A (en) * 1988-04-15 1995-01-03 E. I. Du Pont De Nemours And Company Vitro headful packaging system for cloning DNA fragments as large as 95kb
US5192539A (en) 1988-07-21 1993-03-09 Akzo N.V. Infectious bursal disease virus production in continuous cell lines
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
CA2000048A1 (en) * 1988-10-03 1990-04-03 Edward F. Plow Peptides and antibodies that inhibit integrin-ligand bindin g
US5047335A (en) 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
US5198346A (en) * 1989-01-06 1993-03-30 Protein Engineering Corp. Generation and selection of novel DNA-binding proteins and polypeptides
US5096815A (en) * 1989-01-06 1992-03-17 Protein Engineering Corporation Generation and selection of novel dna-binding proteins and polypeptides
GB8900483D0 (en) 1989-01-10 1989-03-08 Celltech Ltd Recombinant dna method
US5223394A (en) * 1989-04-10 1993-06-29 Biogen, Inc. Recombinant dna molecule comprising lymphocyte function-associated antigen 3 phosphatidylinositol linkage signal sequence
DE3923963A1 (en) 1989-07-20 1991-01-31 Behringwerke Ag MUTEINE OF HUMAN ERYTHROPOETIN, THEIR PRODUCTION AND THEIR USE
US5585362A (en) 1989-08-22 1996-12-17 The Regents Of The University Of Michigan Adenovirus vectors for gene therapy
US5240846A (en) * 1989-08-22 1993-08-31 The Regents Of The University Of Michigan Gene therapy vector for cystic fibrosis
US5332567A (en) * 1989-08-24 1994-07-26 Immunomedics Detection and treatment of infections with immunoconjugates
US5436146A (en) * 1989-09-07 1995-07-25 The Trustees Of Princeton University Helper-free stocks of recombinant adeno-associated virus vectors
US5545522A (en) * 1989-09-22 1996-08-13 Van Gelder; Russell N. Process for amplifying a target polynucleotide sequence using a single primer-promoter complex
US5856298A (en) 1989-10-13 1999-01-05 Amgen Inc. Erythropoietin isoforms
US5670488A (en) * 1992-12-03 1997-09-23 Genzyme Corporation Adenovirus vector for gene therapy
AU7906691A (en) * 1990-05-23 1991-12-10 United States of America, as represented by the Secretary, U.S. Department of Commerce, The Adeno-associated virus (aav)-based eucaryotic vectors
US5349053A (en) * 1990-06-01 1994-09-20 Protein Design Labs, Inc. Chimeric ligand/immunoglobulin molecules and their uses
US5246921A (en) * 1990-06-26 1993-09-21 The Wistar Institute Of Anatomy And Biology Method for treating leukemias
US6743623B2 (en) 1991-09-27 2004-06-01 Centre National De La Recherche Scientifique Viral recombinant vectors for expression in muscle cells
US5521291A (en) * 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
NZ244306A (en) * 1991-09-30 1995-07-26 Boehringer Ingelheim Int Composition for introducing nucleic acid complexes into eucaryotic cells, complex containing nucleic acid and endosomolytic agent, peptide with endosomolytic domain and nucleic acid binding domain and preparation
CA2053187A1 (en) 1991-10-10 1993-04-11 National Research Council Of Canada High level recombinant protein production using conditional helper-free adenovirus vector
US5518913A (en) 1991-10-10 1996-05-21 National Research Council Of Canada High level recombinant protein production using conditional helper-free adenovirus vector
US5384249A (en) 1991-12-17 1995-01-24 Kyowa Hakko Kogyo Co., Ltd. α2→3 sialyltransferase
ES2312164T3 (en) 1992-09-25 2009-02-16 Aventis Pharma S.A. ADENOVIRUS VECTORS FOR THE TRANSFER OF STRANGE GENES IN CELLS OF THE CENTRAL NERVOUS SYSTEM, PARTICULARLY IN THE BRAIN.
ATE307212T1 (en) 1992-11-18 2005-11-15 Arch Dev Corp ADENOVIRUS-DIRECTED GENE TRANSFER TO THE HEART AND VASCULAR SMOOTH MUSCLE
EP1024198A3 (en) * 1992-12-03 2002-05-29 Genzyme Corporation Pseudo-adenoviral vectors for the gene therapy of haemophiliae
DK0604684T3 (en) 1992-12-23 1998-01-26 Campina Melkunie Bv Process for Recovery of Alphalactalbumin and Betalactoglobulin from a Milk Protein Product
DE4311651A1 (en) 1993-04-08 1994-10-13 Boehringer Ingelheim Int Virus for the transport of foreign DNA into higher eukaryotic cells
JPH09500782A (en) * 1993-04-08 1997-01-28 ジェネティック セラピー,インコーポレイテッド Adenovirus vector containing DNA encoding lung surfactant protein
FR2704234B1 (en) 1993-04-22 1995-07-21 Centre Nat Rech Scient RECOMBINANT VIRUSES, PREPARATION AND USE IN GENE THERAPY.
FR2705361B1 (en) * 1993-05-18 1995-08-04 Centre Nat Rech Scient Viral vectors and use in gene therapy.
US6133028A (en) * 1993-05-28 2000-10-17 Transgene S.A. Defective adenoviruses and corresponding complementation lines
US5919676A (en) * 1993-06-24 1999-07-06 Advec, Inc. Adenoviral vector system comprising Cre-loxP recombination
ATE304604T1 (en) * 1993-06-24 2005-09-15 Frank L Graham ADENOVIRUS VECTORS FOR GENE THERAPY
FR2707664B1 (en) 1993-07-13 1995-09-29 Centre Nat Rech Scient Viral vectors and use in gene therapy.
US5543328A (en) * 1993-08-13 1996-08-06 Genetic Therapy, Inc. Adenoviruses having modified fiber proteins
US5552311A (en) * 1993-09-14 1996-09-03 University Of Alabama At Birmingham Research Foundation Purine nucleoside phosphorylase gene therapy for human malignancy
US5534423A (en) * 1993-10-08 1996-07-09 Regents Of The University Of Michigan Methods of increasing rates of infection by directing motion of vectors
US5658763A (en) 1993-10-25 1997-08-19 Creative Biomolecules, Inc. Methods and compositions for high protein production from non-native DNA
US20010006629A1 (en) 1993-10-25 2001-07-05 Richard J. Gregory Recombinant adenoviral vector and methods of use
US6210939B1 (en) * 1993-10-25 2001-04-03 Canji, Inc. Recombinant adenoviral vector and methods of use
US5773569A (en) 1993-11-19 1998-06-30 Affymax Technologies N.V. Compounds and peptides that bind to the erythropoietin receptor
US5830851A (en) 1993-11-19 1998-11-03 Affymax Technologies N.V. Methods of administering peptides that bind to the erythropoietin receptor
US5443953A (en) * 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
WO1995016772A1 (en) 1993-12-14 1995-06-22 Cornell Research Foundation, Inc. Adenovirus gene expression system
US6057299A (en) 1994-01-13 2000-05-02 Calydon, Inc. Tissue-specific enhancer active in prostate
US5698443A (en) 1995-06-27 1997-12-16 Calydon, Inc. Tissue specific viral vectors
CA2117668C (en) 1994-03-09 2005-08-09 Izumu Saito Recombinant adenovirus and process for producing the same
AU2194895A (en) 1994-03-25 1995-10-17 Uab Research Foundation, The Composition and methods for creating syngeneic recombinant virus-producing cells
US5789247A (en) 1994-04-01 1998-08-04 Ballay; Annick Expression in non-tumoral human lymphoblastoid lines with an integrative vector
AU693545B2 (en) 1994-04-26 1998-07-02 Chugai Seiyaku Kabushiki Kaisha Melt granulated sucralfate preparations and a process for their production
US5851806A (en) * 1994-06-10 1998-12-22 Genvec, Inc. Complementary adenoviral systems and cell lines
FR2721943B1 (en) 1994-06-29 1996-08-02 Rhone Poulenc Rorer Sa ADENOVIRUS COMPRISING A GENE ENCODING A SUPEROXIDE DISMUTASE
US6204052B1 (en) * 1994-08-16 2001-03-20 Introgene B.V. Adenoviral vectors with reduced TNF response and partial E3 region deletion
US5559099A (en) * 1994-09-08 1996-09-24 Genvec, Inc. Penton base protein and methods of using same
US5846782A (en) * 1995-11-28 1998-12-08 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
US5552309A (en) * 1994-09-30 1996-09-03 Indiana University Foundation Use of polyols for improving the introduction of genetic material into cells
FR2725726B1 (en) * 1994-10-17 1997-01-03 Centre Nat Rech Scient VIRAL VECTORS AND USE IN GENE THERAPY
FR2726285B1 (en) 1994-10-28 1996-11-29 Centre Nat Rech Scient ADENOVIRUSES CONTAINING VIABLE CONTAMINANT PARTICLES, PREPARATION AND USE
WO1996013597A2 (en) * 1994-10-28 1996-05-09 The Trustees Of The University Of Pennsylvania Improved adenovirus and methods of use thereof
US5856152A (en) * 1994-10-28 1999-01-05 The Trustees Of The University Of Pennsylvania Hybrid adenovirus-AAV vector and methods of use therefor
US5872005A (en) 1994-11-03 1999-02-16 Cell Genesys Inc. Packaging cell lines for adeno-associated viral vectors
CA2206205A1 (en) * 1994-11-28 1996-06-06 Genetic Therapy, Inc. Tissue-specific treatment, diagnostic methods, and compositions using replication-deficient vectors
CA2206683A1 (en) * 1994-12-12 1996-06-20 Genetic Therapy, Inc. Improved adenoviral vectors and producer cells
FR2727867B1 (en) 1994-12-13 1997-01-31 Rhone Poulenc Rorer Sa GENE TRANSFER IN MEDULLAR MOTONURONES USING ADENOVIRAL VECTORS
GB9500659D0 (en) 1995-01-13 1995-03-08 Camco Drilling Group Ltd Improvements in or relating to rotary drill bits
US6127525A (en) * 1995-02-21 2000-10-03 Cornell Research Foundation, Inc. Chimeric adenoviral coat protein and methods of using same
US5770442A (en) * 1995-02-21 1998-06-23 Cornell Research Foundation, Inc. Chimeric adenoviral fiber protein and methods of using same
US5652224A (en) * 1995-02-24 1997-07-29 The Trustees Of The University Of Pennsylvania Methods and compositions for gene therapy for the treatment of defects in lipoprotein metabolism
US6037453A (en) 1995-03-15 2000-03-14 Genentech, Inc. Immunoglobulin variants
US5707618A (en) * 1995-03-24 1998-01-13 Genzyme Corporation Adenovirus vectors for gene therapy
ATE243260T1 (en) * 1995-04-17 2003-07-15 Univ Texas ADENOVIRUS HELPER VIRUS SYSTEM
JPH08303576A (en) 1995-05-10 1996-11-19 Aisin Seiki Co Ltd Shift control device for automatic transmission
US5767078A (en) 1995-06-07 1998-06-16 Johnson; Dana L. Agonist peptide dimers
US6783980B2 (en) 1995-06-15 2004-08-31 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
US6265212B1 (en) 1995-06-15 2001-07-24 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
ATE445705T1 (en) * 1995-06-15 2009-10-15 Crucell Holland Bv PACKAGING SYSTEMS FOR HUMAN RECOMBINANT ADENOVIRUSES FOR GENE THERAPY
FR2735789B1 (en) * 1995-06-23 1997-07-25 Centre Nat Rech Scient RECOMBINANT ADENOVIRUSES, THEIR USE FOR PREPARING AVA, COMPLEMENTARY CELL LINE AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US6676935B2 (en) 1995-06-27 2004-01-13 Cell Genesys, Inc. Tissue specific adenoviral vectors
US7001764B2 (en) 1995-06-27 2006-02-21 Cell Genesys, Inc. Compositions comprising tissue specific adenoviral vectors
FR2737222B1 (en) * 1995-07-24 1997-10-17 Transgene Sa NEW VIRAL AND LINEAR VECTORS FOR GENE THERAPY
FR2737501B1 (en) * 1995-07-31 1997-10-24 Transgene Sa NOVEL AUXILIARY VIRUSES FOR THE PREPARATION OF RECOMBINANT VIRAL VECTORS
US5622699A (en) * 1995-09-11 1997-04-22 La Jolla Cancer Research Foundation Method of identifying molecules that home to a selected organ in vivo
US5837511A (en) 1995-10-02 1998-11-17 Cornell Research Foundation, Inc. Non-group C adenoviral vectors
EA003195B1 (en) 1995-11-13 2003-02-27 Такара Сузо Ко., Лтд. Method for gene transfer into target cells with retrovirus
DK0870044T3 (en) * 1995-12-01 2004-08-02 Crucell Holland Bv Regulated protein expression in stably transfected mammalian cells
US5871727A (en) * 1995-12-08 1999-02-16 Uab Research Foundation Targeted adenovirus vectors
AU731615B2 (en) * 1996-02-09 2001-04-05 Het Nederlands Kanker Instituut Vectors and methods for providing cells with additional nucleic acid material integrated in the genome of said cells
US5835382A (en) 1996-04-26 1998-11-10 The Scripps Research Institute Small molecule mimetics of erythropoietin
US5891690A (en) * 1996-04-26 1999-04-06 Massie; Bernard Adenovirus E1-complementing cell lines
US5877011A (en) * 1996-11-20 1999-03-02 Genzyme Corporation Chimeric adenoviral vectors
US5922315A (en) * 1997-01-24 1999-07-13 Genetic Therapy, Inc. Adenoviruses having altered hexon proteins
EP0856585A1 (en) * 1997-01-29 1998-08-05 Introgene B.V. A conditional replication and expression system
WO1999066041A1 (en) 1998-06-16 1999-12-23 Human Genome Sciences, Inc. 94 human secreted proteins
WO1998039035A1 (en) * 1997-03-07 1998-09-11 The Wistar Institute Of Anatomy & Biology Method and compositions for healing tissue defects and inducing hypervascularity in mammalian tissue
CA2221819A1 (en) 1997-03-27 1998-09-27 Thomas A. Gigliatti Insect expression vectors
US5981275A (en) 1997-04-14 1999-11-09 Genzyme Corporation Transgene expression system for increased persistence
US6100086A (en) 1997-04-14 2000-08-08 Genzyme Corporation Transgene expression systems
AU743051B2 (en) 1997-05-08 2002-01-17 Genetic Therapy, Inc. Gene transfer with adenoviruses having modified fiber proteins
US5849561A (en) * 1997-05-22 1998-12-15 Cornell Research Foundation, Inc. Method for the production of non-group C adenoviral vectors
US6287857B1 (en) * 1998-02-09 2001-09-11 Genzyme Corporation Nucleic acid delivery vehicles
US6417168B1 (en) 1998-03-04 2002-07-09 The Trustees Of The University Of Pennsylvania Compositions and methods of treating tumors
WO1999047180A1 (en) 1998-03-20 1999-09-23 Genzyme Corporation Chimeric adenoviral vectors for targeted gene delivery
US6670188B1 (en) 1998-04-24 2003-12-30 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
US6413776B1 (en) * 1998-06-12 2002-07-02 Galapagos Geonomics N.V. High throughput screening of gene function using adenoviral libraries for functional genomics applications
US20030017138A1 (en) 1998-07-08 2003-01-23 Menzo Havenga Chimeric adenoviruses
EP1020529B1 (en) 1998-11-20 2005-06-01 Crucell Holland B.V. Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US6929946B1 (en) 1998-11-20 2005-08-16 Crucell Holland B.V. Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
EP1016726A1 (en) 1998-12-30 2000-07-05 Introgene B.V. Gene therapy to promote angiogenesis
US6855544B1 (en) 1999-04-15 2005-02-15 Crucell Holland B.V. Recombinant protein production in a human cell
WO2003048197A1 (en) 2001-12-07 2003-06-12 Crucell Holland B.V. Production and of viruses, viral isolates and vaccines
ATE364707T1 (en) 1999-05-17 2007-07-15 Crucell Holland Bv RECOMBINANT HUMAN ADENOVIRUS SEROTYPE 35
US6492169B1 (en) 1999-05-18 2002-12-10 Crucell Holland, B.V. Complementing cell lines
US20050232900A1 (en) 1999-05-18 2005-10-20 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US6913922B1 (en) 1999-05-18 2005-07-05 Crucell Holland B.V. Serotype of adenovirus and uses thereof
EP1067188A1 (en) 1999-07-08 2001-01-10 Introgene B.V. Infection with chimaeric adenoviruses of cells negative for the adenovirus serotype 5 Coxsacki adenovirus receptor (CAR)
US6558948B1 (en) 1999-11-23 2003-05-06 Stefan Kochanek Permanent amniocytic cell line, its production and use for the production of gene transfer vectors
US7521220B2 (en) 1999-11-26 2009-04-21 Crucell Holland B.V. Production of vaccines
EP1103610A1 (en) 1999-11-26 2001-05-30 Introgene B.V. Production of vaccines from immortalised mammalian cell lines
US7527961B2 (en) 1999-11-26 2009-05-05 Crucell Holland B.V. Production of vaccines
US7125705B2 (en) 2000-04-28 2006-10-24 Genzyme Corporation Polynucleotides for use in recombinant adeno-associated virus virion production
EP1157999A1 (en) 2000-05-24 2001-11-28 Introgene B.V. Methods and means for enhancing skin transplantation using gene delivery vehicles having tropism for primary fibroblasts, as well as other uses thereof
US6844192B2 (en) 2001-06-29 2005-01-18 Wake Forest University Adenovirus E4 protein variants for virus production
US20030026783A1 (en) 2001-08-03 2003-02-06 Amine Abina Method of modulating neutralizing antibodies formation in mammals, and uses thereof in gene therapy, animal transgenesis and in functional inactivation of an endogenous proteins
JP4583029B2 (en) 2001-10-29 2010-11-17 クルセル ホランド ベー ヴェー Method and method for producing a protein having a predetermined post-translational modification
CN100547069C (en) 2001-12-07 2009-10-07 克鲁塞尔荷兰公司 The production of virus, viral isolates and vaccine
IL164802A0 (en) 2002-04-25 2005-12-18 Crucell Holland Bv Stable adenoviral vectors and methods for propagation thereof
EA010828B1 (en) 2002-04-25 2008-12-30 Круселл Холланд Б.В. Recombinant adenovirus vectors and methods for the production thereof and use
SI1523496T1 (en) 2002-07-18 2011-11-30 Merus B V Recombinant production of mixtures of antibodies
AU2003288273A1 (en) 2002-10-23 2004-05-13 Crucell Holland B.V. New settings for recombinant adenoviral-based vaccines
EP1573012B1 (en) 2002-12-17 2011-11-30 Crucell Holland B.V. Recombinant viral-based malaria vaccines
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
JP2007522814A (en) 2004-02-23 2007-08-16 クルセル ホランド ベー ヴェー Virus purification method
US20080131461A1 (en) 2004-10-14 2008-06-05 Crucell Holland B.V. Malaria Prime/Boost Vaccines
US20070010016A1 (en) 2005-03-11 2007-01-11 Mccelland Alan Gene transfer with adenoviruses having modified fiber proteins

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994028152A1 (en) * 1993-05-28 1994-12-08 Transgene S.A. Defective adenoviruses and corresponding complementation lines
WO1995002697A1 (en) * 1993-07-13 1995-01-26 Rhone-Poulenc Rorer S.A. Defective adenovirus vectors and use thereof in gene therapy
WO1995027071A2 (en) * 1994-04-04 1995-10-12 Board Of Regents, The University Of Texas System An adenovirus supervector system
WO1995034671A1 (en) * 1994-06-10 1995-12-21 Genvec, Inc. Complementary adenoviral vector systems and cell lines

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BROUGH ET AL.: "Restricted changes in the adenovirus DNA-binding protein that lead to extended host range or temperature-sensitive phenotypes", JOURNAL OF VIROLOGY, vol. 55, pages 206 - 212, XP002012304 *
ENGELHARDT ET AL.: "Ablation of E2A in recombinant adenoviruses improves transgene persistence and decreases inflammatory response in mouse liver", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, vol. 91, WASHINGTON US, pages 6196 - 6200, XP002012303 *

Cited By (221)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6472212B1 (en) 1991-10-04 2002-10-29 Introgene B.V. Methods and compositions for genetically modifying primate bone marrow cells
US6204052B1 (en) 1994-08-16 2001-03-20 Introgene B.V. Adenoviral vectors with reduced TNF response and partial E3 region deletion
US6395519B1 (en) 1995-06-15 2002-05-28 Introgene B.V. Means and methods for nucleic acid delivery vehicle design and nucleic acid transfer
US6692966B2 (en) 1995-06-15 2004-02-17 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
US6265212B1 (en) 1995-06-15 2001-07-24 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
US6261554B1 (en) 1995-07-25 2001-07-17 Introgene B.V. Compositions for targeted gene delivery
US7033834B2 (en) 1995-07-25 2006-04-25 Crucell Holland B.V. Methods and means for targeted gene delivery
US7781208B2 (en) 1995-07-25 2010-08-24 Crucell Holland B.V. Methods and means for targeted gene delivery
US6140103A (en) * 1995-12-01 2000-10-31 Introgene B.V. Regulated protein expression in stably transfected mammalian cells
US6312957B1 (en) 1996-12-05 2001-11-06 Introgene B.V. Genetic modification of primate hemopoietic repopulating stem cells
EP0973866A1 (en) 1997-03-04 2000-01-26 Baxter International Inc. Adenovirus e1-complementing cell lines
FR2774699A1 (en) * 1997-11-17 1999-08-13 Rhone Poulenc Rorer Sa METHOD FOR REDUCING HOMOLOGATED RECOMBINATION EVENTS
WO1999025861A2 (en) * 1997-11-17 1999-05-27 Aventis Pharma S.A. Adenovirus vectors and method for reducing homologous recombination phenomena
US6410298B1 (en) 1997-11-17 2002-06-25 Aventis Pharma S.A. Adenovirus vectors and method for reducing homologous recombination phenomena
CZ299797B6 (en) * 1997-11-17 2008-11-26 Gencell Sa Method of reducing homologous recombination phenomena, adenoviral vectors and cells containing defective adenovirus
WO1999025861A3 (en) * 1997-11-17 1999-07-01 Rhone Poulenc Rorer Sa Adenovirus vectors and method for reducing homologous recombination phenomena
WO1999027123A3 (en) * 1997-11-26 1999-08-12 Univ Texas Modified sv40 viral vectors
WO1999027123A2 (en) * 1997-11-26 1999-06-03 Board Of Regents, The University Of Texas System Modified sv40 viral vectors
US6468771B1 (en) 1997-12-23 2002-10-22 Introgene Adeno-associated virus and adenovirus chimeric recombinant viruses useful for the integration of foreign genetic information into the chromosomal DNA of target cells
WO1999032647A1 (en) * 1997-12-23 1999-07-01 Introgene B.V. Adeno-associated virus and adenovirus chimeric recombinant viruses useful for the integration of foreign genetic information into the chromosomal dna of target cells
US6803194B1 (en) 1998-02-13 2004-10-12 Hk Pharmaceuticals, Inc. Use of ribozymes for functionating genes
US5981225A (en) * 1998-04-16 1999-11-09 Baylor College Of Medicine Gene transfer vector, recombinant adenovirus particles containing the same, method for producing the same and method of use of the same
US6670188B1 (en) 1998-04-24 2003-12-30 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
US7037716B2 (en) 1998-04-24 2006-05-02 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
EP0955373A3 (en) * 1998-04-24 2000-04-19 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
US6878549B1 (en) 1998-04-24 2005-04-12 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
WO1999055132A3 (en) * 1998-04-24 2000-04-06 Introgene Bv Generation of packaging system for human recombinant adenoviral vectors
EP0955373A2 (en) * 1998-04-24 1999-11-10 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
WO1999055132A2 (en) * 1998-04-24 1999-11-04 Introgene B.V. Generation of packaging system for human recombinant adenoviral vectors
US6498027B1 (en) 1998-05-20 2002-12-24 Introgene B.V. Targeted delivery through a cationic amino acid transporter
WO1999064582A3 (en) * 1998-06-12 2000-04-06 Introgene Bv High-throughput screening of gene function using libraries for functional genomics applications
US6340595B1 (en) 1998-06-12 2002-01-22 Galapagos Genomics N.V. High throughput screening of gene function using adenoviral libraries for functional genomics applications
AU756605B2 (en) * 1998-06-12 2003-01-16 Galapagos N.V. High-throughput screening of gene function using libraries for functional genomics applications
US7029848B2 (en) 1998-06-12 2006-04-18 Galapagos Genomics N.V. High throughput screening of gene function using libraries for functional genomics applications
WO1999064582A2 (en) * 1998-06-12 1999-12-16 Introgene B.V. High-throughput screening of gene function using libraries for functional genomics applications
EP1022335A1 (en) * 1998-06-12 2000-07-26 Introgene B.V. High-throughput screening of gene function using libraries for functional genomics applications
US6413776B1 (en) 1998-06-12 2002-07-02 Galapagos Geonomics N.V. High throughput screening of gene function using adenoviral libraries for functional genomics applications
US7749493B2 (en) 1998-07-08 2010-07-06 Crucell Holland B.V. Chimeric adenoviruses
WO2000024916A1 (en) * 1998-10-27 2000-05-04 Crucell Holland B.V. Improved aav vector production
AU768489B2 (en) * 1998-11-06 2003-12-11 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Cells for the production of helper dependent adenoviral vectors, method for the preparation and use thereof
JP2000157289A (en) * 1998-11-20 2000-06-13 Introgene Bv Gene delivery vector provided with tissue tropism to smooth muscle cell and/or endothelial cell
JP4683682B2 (en) * 1998-11-20 2011-05-18 クルーセル ホランド ベスローテン フェンノートシャップ Gene delivery vector imparted tissue tropism to smooth muscle cells and / or endothelial cells
US6929946B1 (en) 1998-11-20 2005-08-16 Crucell Holland B.V. Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US6447768B1 (en) * 1998-12-30 2002-09-10 Introgene B.V. Methods of gene therapy with a DNA sequence encoding NOS
US6869936B1 (en) 1999-03-04 2005-03-22 Crucell Holland B.V. Means and methods for fibroblast-like or macrophage-like cell transduction
JP4683727B2 (en) * 1999-03-04 2011-05-18 クルーセル ホランド ベスローテン フェンノートシャップ Means and methods for transduction of fibroblast-like or macrophage-like cells
US7238528B2 (en) 1999-03-04 2007-07-03 Crucell Holland B.V. Means and methods for fibroblast-like or macrophage-like cell transduction
JP2002537816A (en) * 1999-03-04 2002-11-12 イントロヘーネ ベスローテン フェンノートシャップ Means and methods for transducing fibroblast-like or macrophage-like cells
US7491532B2 (en) 1999-04-15 2009-02-17 Crucell Holland, B.V. Recombinant protein production in a human cell
US6855544B1 (en) 1999-04-15 2005-02-15 Crucell Holland B.V. Recombinant protein production in a human cell
WO2000063403A2 (en) * 1999-04-15 2000-10-26 Crucell Holland B.V. Recombinant protein production in a human cell using sequences encoding adenovirus e1 protein
US7470523B2 (en) 1999-04-15 2008-12-30 Crucell Holland B.V. Recombinant protein production in a human cell
CN100457914C (en) * 1999-04-15 2009-02-04 荷兰克鲁塞尔公司 Recombinant protein production in human cell using sequences encoding adenovirus E1 protein
US7604960B2 (en) 1999-04-15 2009-10-20 Crucell Holland B.V. Transient protein expression methods
US7132280B2 (en) 1999-04-15 2006-11-07 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
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
EP1161548B2 (en) 1999-04-15 2009-10-14 Crucell Holland B.V. Recombinant protein production in a human cell using sequences encoding adenovirus e1 protein
EP1533380A2 (en) 1999-04-15 2005-05-25 Crucell Holland B.V. Recombinant protein production in a human cell comprising at least one E1 protein of adenovirus
EP2163640A1 (en) 1999-04-15 2010-03-17 Crucell Holland B.V. Recombinant protein production in a human cell comprising at least one E1 protein of adenovirus
EP1533380A3 (en) * 1999-04-15 2005-06-22 Crucell Holland B.V. Recombinant protein production in a human cell comprising at least one E1 protein of adenovirus
US7833753B2 (en) 1999-04-15 2010-11-16 Crucell Holland B.V. Methods of producing erythropoietin isoforms comprising Lewis-X structures and high sialic acid content and compositions of the same
WO2000063403A3 (en) * 1999-04-15 2001-02-15 Introgene Bv Recombinant protein production in a human cell using sequences encoding adenovirus e1 protein
WO2001007571A1 (en) * 1999-04-23 2001-02-01 Introgene B.V. Means and methods for nucleic acid transfer
US6913922B1 (en) 1999-05-18 2005-07-05 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US7250293B2 (en) 1999-05-18 2007-07-31 Crucell Holland B.V. Complementing cell lines
US6787351B2 (en) 1999-07-06 2004-09-07 Merck & Co., Inc. Adenovirus carrying gag gene HIV vaccine
EP1083228A1 (en) * 1999-09-10 2001-03-14 Introgene B.V. Modified adenoviral vectors for use in gene therapy
WO2001020014A1 (en) * 1999-09-10 2001-03-22 Crucell Holland B.V. Modified adenoviral vectors for use in gene therapy
EP1083229A1 (en) * 1999-09-10 2001-03-14 Introgene B.V. Modified adenoviral vectors for use in gene therapy
US6365394B1 (en) 1999-09-29 2002-04-02 The Trustees Of The University Of Pennsylvania Cell lines and constructs useful in production of E1-deleted adenoviruses in absence of replication competent adenovirus
WO2001023597A2 (en) * 1999-09-29 2001-04-05 The Trustees Of The University Of Pennsylvania Cell lines and constructs useful in production of e1-deleted adenoviruses in absence of replication competent adenovirus
WO2001023597A3 (en) * 1999-09-29 2001-10-25 Univ Pennsylvania Cell lines and constructs useful in production of e1-deleted adenoviruses in absence of replication competent adenovirus
US7115391B1 (en) 1999-10-01 2006-10-03 Genovo, Inc. Production of recombinant AAV using adenovirus comprising AAV rep/cap genes
CZ300124B6 (en) * 1999-11-18 2009-02-18 Cevec Pharmaceuticals Gmbh Permanent cell line of amniocytes, process of its preparation and its use for producing vectors for transfer of genes
WO2001036615A3 (en) * 1999-11-18 2002-01-10 Stefan Kochanek Permanent amniocyte cell line, the production thereof and its use for producing gene transfer vectors
WO2001036615A2 (en) * 1999-11-18 2001-05-25 Stefan Kochanek Permanent amniocyte cell line, the production thereof and its use for producing gene transfer vectors
CN100412201C (en) * 1999-11-18 2008-08-20 赛维科药物有限公司 Permanent amniocyte cell line, the production thereof and its use for producing gene transfer vectors
US6558948B1 (en) 1999-11-23 2003-05-06 Stefan Kochanek Permanent amniocytic cell line, its production and use for the production of gene transfer vectors
US7833788B2 (en) 1999-11-26 2010-11-16 Crucell Holland B.V. Production of vaccines
US7527961B2 (en) 1999-11-26 2009-05-05 Crucell Holland B.V. Production of vaccines
US7550284B2 (en) 1999-11-26 2009-06-23 Crucell Holland B.V. Production of vaccines
US7521220B2 (en) 1999-11-26 2009-04-21 Crucell Holland B.V. Production of vaccines
US7964198B2 (en) 1999-11-26 2011-06-21 Crucell Holland B.V. Production of vaccines
US7192759B1 (en) 1999-11-26 2007-03-20 Crucell Holland B.V. Production of vaccines
US7132277B1 (en) 2000-01-31 2006-11-07 Merck & Co., Inc. Helper dependent vector system for gene therapy
US6733993B2 (en) 2000-09-15 2004-05-11 Merck & Co., Inc. Enhanced first generation adenovirus vaccines expressing codon optimized HIV1-gag, pol, nef and modifications
US7235233B2 (en) 2000-09-26 2007-06-26 Crucell Holland B.V. Serotype 5 adenoviral vectors with chimeric fibers for gene delivery in skeletal muscle cells or myoblasts
US6908762B2 (en) 2000-10-10 2005-06-21 Genvec, Inc. Method of preparing a eukaryotic viral vector
WO2002031169A1 (en) * 2000-10-10 2002-04-18 Genvec, Inc. Method of preparing a eukaryotic viral vector
US6573092B1 (en) 2000-10-10 2003-06-03 Genvec, Inc. Method of preparing a eukaryotic viral vector
AU2002218569B2 (en) * 2000-11-15 2007-04-05 Crucell Holland B.V. Complementing cell lines
US9228205B2 (en) 2000-11-15 2016-01-05 Crucell Holland B.V. Complementing cell lines
WO2002040665A2 (en) * 2000-11-15 2002-05-23 Crucell Holland B.V. Complementing cell lines
WO2002040665A3 (en) * 2000-11-15 2002-10-17 Crucell Holland Bv Complementing cell lines
US7163787B2 (en) 2001-05-07 2007-01-16 Crucell Holland B.V. Methods for the identification of antiviral compounds
US6682929B2 (en) 2001-07-23 2004-01-27 Genvec, Inc. Adenovector complementing cells
US7504248B2 (en) 2001-12-07 2009-03-17 Crucell Holland B.V. Production of viruses viral isolates and vaccines
US8802417B2 (en) 2001-12-07 2014-08-12 Crucell Holland B.V. Production of viruses, viral isolates and vaccines
EP3483183A1 (en) 2002-03-01 2019-05-15 Immunomedics, Inc. Immunoconjugate comprising humanised rs7 antibodies
EP2287201A2 (en) 2002-03-01 2011-02-23 Immunomedics, Inc. RS7 antibodies
EP2301968A2 (en) 2002-06-14 2011-03-30 Immunomedics, Inc. Humanized monoclonal antibody HPAM4
US7524947B2 (en) 2002-12-17 2009-04-28 Crucell Holland B.V. Recombinant viral-based malaria vaccines
US7521229B2 (en) 2002-12-17 2009-04-21 Crucell Holland B.V. Recombinant viral-based malaria vaccines
US8097453B2 (en) 2002-12-17 2012-01-17 Crucell Holland B.V. Recombinant viral-based malaria vaccines
US8361478B2 (en) 2002-12-17 2013-01-29 Crucell Holland B.V. Recombinant viral-based malaria vaccines
US7387894B2 (en) 2002-12-17 2008-06-17 Crucell Holland B.V. Recombinant viral-based malaria vaccines
US7867764B2 (en) 2002-12-17 2011-01-11 Crucell Holland B.V. Recombinant viral-based malaria vaccines
EP2258850A1 (en) 2002-12-17 2010-12-08 Crucell Holland B.V. Recombinant viral-based malaria vaccines
US7300657B2 (en) 2002-12-17 2007-11-27 Crucell Holland B.V. Recombinant viral-based malaria vaccines
EP2824185A1 (en) 2003-02-24 2015-01-14 GenVec, Inc. Materials and methods for treating disorders of the ear
US7608431B2 (en) 2003-05-09 2009-10-27 Crucell Holland B.V. Fed-batch process for production of erythropoietin in human embryonic retina cells that express adenovirus E1A
US7291484B2 (en) 2003-05-09 2007-11-06 Crucell Holland B.V. Processes for culturing E1-immortalized cells to increase product yields
US8008043B2 (en) 2003-05-09 2011-08-30 Crucell Holland B.V. Cultures of E1-immortalized cells and processes for culturing the same to increase product yields therefrom
EP1644491A4 (en) * 2003-07-03 2007-01-03 Cell Genesys Inc Adenoviral e1a/e1b complementing cell line
EP1644491A2 (en) * 2003-07-03 2006-04-12 Cell Genesys, Inc. Adenoviral e1a/e1b complementing cell line
US8940534B2 (en) 2003-11-03 2015-01-27 Probiogen Ag Immortalized avian cell lines for virus production
EP2168603A1 (en) 2003-11-14 2010-03-31 GenVec, Inc. Therapeutic regimen for treating cancer
EP2163260A2 (en) 2004-01-23 2010-03-17 Istituto di Richerche di Biologia Molecolare P. Angeletti S.p.A. Chimpanzee adenovirus vaccine carriers
US8673319B2 (en) 2004-01-23 2014-03-18 Msd Italia Srl Chimpanzee adenovirus vaccine carriers
US8216834B2 (en) 2004-01-23 2012-07-10 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Chimpanzee adenovirus vaccine carriers
EP3269390A2 (en) 2004-01-23 2018-01-17 MSD Italia S.r.l. Chimpanzee adenovirus vaccine carriers
EP2567967A2 (en) 2004-04-12 2013-03-13 GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Method of using adenoviral vectors to induce an immune response
WO2006031689A2 (en) 2004-09-13 2006-03-23 Genzyme Corporation Multimeric constructs
EP2229956A1 (en) 2004-09-13 2010-09-22 Genzyme Corporation Multimeric constructs
US8309349B2 (en) 2004-10-28 2012-11-13 University of Pittsburgh—of the Commonwealth System of Higher Education Cell line
US8765146B2 (en) 2005-08-31 2014-07-01 Genvec, Inc. Adenoviral vector-based malaria vaccines
US8323663B2 (en) 2005-11-10 2012-12-04 Genvec, Inc. Adenoviral vector-based foot-and-mouth disease vaccine
US10080783B2 (en) 2006-01-20 2018-09-25 Genzyme Corporation Intraventricular enzyme delivery for lysosomal storage diseases
US11253485B2 (en) 2006-02-09 2022-02-22 Genzyme Corporation Slow intraventricular delivery
EP3459441A1 (en) 2006-02-09 2019-03-27 Genzyme Corporation Slow intraventricular delivery
WO2007095056A2 (en) 2006-02-09 2007-08-23 Genzyme Corporation Slow intraventricular delivery
WO2008112540A2 (en) * 2007-03-09 2008-09-18 Vectorlogics, Inc. Cells for adenovirus vector and protein production
WO2008112540A3 (en) * 2007-03-09 2008-12-04 Vectorlogics Inc Cells for adenovirus vector and protein production
US10995128B2 (en) 2008-04-16 2021-05-04 Ferring International Center Sa Pharmaceutical preparation
EP3045471B1 (en) 2008-04-16 2017-03-29 Ferring B.V. Recombinant fsh including alpha 2,3- and alpha 2,6-sialylation
EP3144318B1 (en) 2008-04-16 2020-09-23 Ferring B.V. Recombinant fsh including alpha 2,3- and alpha 2,6-sialylation
WO2010084100A1 (en) 2009-01-20 2010-07-29 Transgene Sa Soluble icam-1 as biomarker for prediction of therapeutic response
WO2010108908A1 (en) 2009-03-24 2010-09-30 Transgene Sa Biomarker for monitoring patients
WO2010119003A1 (en) 2009-04-17 2010-10-21 Transgene Sa Biomarker for monitoring patients
WO2011003615A2 (en) 2009-07-08 2011-01-13 Glycotope Gmbh Perfusion bioreactor
WO2011003905A1 (en) 2009-07-10 2011-01-13 Transgene Sa Biomarker for selecting patients and related methods
US8546123B2 (en) 2009-07-16 2013-10-01 Crucell Holland B.V. Production of poliovirus at high titers for vaccine production
US9022240B2 (en) 2009-07-16 2015-05-05 Crucell Holland B.V. Production of poliovirus at high titers for vaccine production
WO2011015656A2 (en) 2009-08-07 2011-02-10 Transgene Sa Composition for treating hbv infection
WO2011045381A1 (en) * 2009-10-15 2011-04-21 Crucell Holland B.V. Process for adenovirus purification from high cell density cultures
EA028623B1 (en) * 2009-10-15 2017-12-29 Янссен Вэксинс Энд Превеншн Б.В. Method for the purification of adenovirus particles
US8470585B2 (en) 2009-10-15 2013-06-25 Crucell Holland B.V. Process for adenovirus purification from high cell density cultures
US8460920B2 (en) 2009-10-15 2013-06-11 Crucell Holland B.V. Method for the purification of adenovirus particles
WO2011045378A1 (en) * 2009-10-15 2011-04-21 Crucell Holland B.V. Method for the purification of adenovirus particles
EA028875B1 (en) * 2009-10-15 2018-01-31 Янссен Вэксинс Энд Превеншн Б.В. Method for purifying adenovirus particles from a cell suspension
WO2011047316A1 (en) 2009-10-16 2011-04-21 The United States Of America, As Represented By The Secretary Department Of Health And Human Services Nucleic acid sequences encoding expandable hiv mosaic proteins
EP2853266A1 (en) 2009-11-09 2015-04-01 Genvec, Inc. Methods of propagating monkey adenoviral vectors
WO2011057254A2 (en) 2009-11-09 2011-05-12 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Simian adenoviral vector-based vaccines
WO2011057248A2 (en) 2009-11-09 2011-05-12 Genvec, Inc. Simian adenovirus and methods of use
WO2011116189A1 (en) 2010-03-17 2011-09-22 Cornell University Disrupted adenovirus-based vaccine against drugs of abuse
EP3284821A1 (en) 2010-03-23 2018-02-21 Intrexon Corporation Vectors conditionally expressing therapeutic proteins, host cells comprising the vectors, and uses thereof
WO2012021730A2 (en) 2010-08-11 2012-02-16 Genvec, Inc. Respiratory syncytial virus (rsv) vaccine
WO2012083297A2 (en) 2010-12-17 2012-06-21 Genvec, Inc. Adenoviral vectors with modified hexon regions
WO2012088041A1 (en) 2010-12-20 2012-06-28 Genvec, Inc. Adenoviral vector-based dengue fever vaccine
WO2012122025A2 (en) 2011-03-04 2012-09-13 Intrexon Corporation Vectors conditionally expressing protein
EP3450568A2 (en) 2011-03-04 2019-03-06 Intrexon Corporation Vectors conditionally expressing protein
WO2013007772A1 (en) 2011-07-12 2013-01-17 Transgene Sa Hbv polymerase mutants
WO2013045658A1 (en) 2011-09-29 2013-04-04 Transgene Sa Immunotherapy composition and regimen for treating hepatitis c virus infection
WO2013045668A2 (en) 2011-09-29 2013-04-04 Transgene Sa Immunotherapy composition and regimen for treating hepatitis c virus infection
WO2013052832A2 (en) 2011-10-05 2013-04-11 Genvec, Inc. Adenoviral vectors and methods of use
US9233153B2 (en) 2011-10-05 2016-01-12 Genvec, Inc. Affenadenovirus (gorilla) or adenoviral vectors and methods of use
WO2013052859A2 (en) 2011-10-05 2013-04-11 Genvec, Inc. Adenoviral vector-based respiratory syncytial virus (rsv) vaccine
US10272162B2 (en) 2011-10-05 2019-04-30 Genvec, Inc. Affenadenovirus (gorilla) or adenoviral vectors and methods of use
US10260074B2 (en) 2011-10-05 2019-04-16 Genvec, Inc. Affenadenovirus (gorilla) or adenoviral vectors and methods of use
US9580476B2 (en) 2011-10-05 2017-02-28 Genvec, Inc. Adenoviral vector-based respiratory syncytial virus (RSV) vaccine
WO2013052799A2 (en) 2011-10-05 2013-04-11 Genvec, Inc. Adenoviral vectors and methods of use
US9617560B2 (en) 2011-10-05 2017-04-11 Genvec, Inc. Simian (gorilla) adenovirus or adenoviral vectors and methods of use
US9629906B2 (en) 2011-10-05 2017-04-25 Genvec, Inc. Affenadenovirus (gorilla) or adenoviral vectors and methods of use
US10787682B2 (en) 2011-10-05 2020-09-29 Genvec, Inc. Simian (gorilla) adenovirus or adenoviral vectors and methods of use
US9725738B2 (en) 2011-10-05 2017-08-08 Genvec, Inc. Affenadenovirus (gorilla) or adenoviral vectors and methods of use
US10792376B2 (en) 2011-10-05 2020-10-06 Genvec, Inc. Affenadenovirus (gorilla) or adenoviral vectors and methods of use
US10059962B2 (en) 2011-10-05 2018-08-28 Genvec, Inc. Simian (gorilla) adenovirus or adenoviral vectors and methods of use
US11034975B2 (en) 2011-10-05 2021-06-15 Genvec, Inc. Affenadenovirus (gorilla) or adenoviral vectors and methods of use
US11359214B2 (en) 2011-10-05 2022-06-14 Genvec, Inc. Simian (gorilla) adenovirus or adenoviral vectors and methods of use
WO2013052811A2 (en) 2011-10-05 2013-04-11 Genvec, Inc. Adenoviral vectors and methods of use
WO2013116591A1 (en) 2012-02-02 2013-08-08 Genvec, Inc. Adenoviral vector-based malaria vaccine
WO2013119880A1 (en) 2012-02-07 2013-08-15 Global Bio Therapeutics Usa, Inc. Compartmentalized method of nucleic acid delivery and compositions and uses thereof
US9821114B2 (en) 2012-02-07 2017-11-21 Global Bio Therapeutics, Inc. Compartmentalized method of nucleic acid delivery and compositions and uses thereof
EP3970734A2 (en) 2012-02-07 2022-03-23 Global Bio Therapeutics, Inc. Compartmentalized method of nucleic acid delivery and compositions and uses thereof
WO2013181128A1 (en) 2012-05-29 2013-12-05 Genvec, Inc. Modified serotype 28 adenoviral vectors
WO2013180967A1 (en) 2012-05-29 2013-12-05 Genvec, Inc. Herpes simplex virus vaccine
US9676824B2 (en) 2012-05-29 2017-06-13 Genvec, Inc. Herpes simplex virus vaccine
US10125174B2 (en) 2012-05-29 2018-11-13 Genvec, Inc. Herpes simplex virus vaccine
WO2014009433A1 (en) 2012-07-10 2014-01-16 Transgene Sa Mycobacterium resuscitation promoting factor for use as adjuvant
WO2014009438A2 (en) 2012-07-10 2014-01-16 Transgene Sa Mycobacterial antigen vaccine
EP3564380A1 (en) 2012-11-16 2019-11-06 Beth Israel Deaconess Medical Center, Inc. Recombinant adenoviruses and use thereof
US10781427B2 (en) 2012-11-16 2020-09-22 Beth Israel Deaconess Medical Center, Inc. Recombinant adenoviruses and use thereof
WO2014078688A2 (en) 2012-11-16 2014-05-22 Beth Israel Deaconess Medical Center, Inc. Recombinant adenoviruses and use thereof
US10106781B2 (en) 2012-11-16 2018-10-23 Beth Israel Deaconess Medical Center, Inc. Recombinant adenoviruses and use thereof
ES2525768A1 (en) * 2013-06-24 2014-12-29 Curaxis, S.L. Procedure for the production of glycosylated recombinant proteins. (Machine-translation by Google Translate, not legally binding)
US11364032B2 (en) 2013-08-08 2022-06-21 Global Bio Therapeutics, Inc. Clamp device for minimally invasive procedures and uses thereof
WO2015104380A1 (en) 2014-01-09 2015-07-16 Transgene Sa Fusion of heterooligomeric mycobacterial antigens
WO2016037164A1 (en) 2014-09-07 2016-03-10 Selecta Biosciences, Inc. Methods and compositions for attenuating gene expression modulating anti-viral transfer vector immune responses
WO2016037162A1 (en) 2014-09-07 2016-03-10 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector immune responses
US11279951B2 (en) 2014-10-09 2022-03-22 Genvec, Inc. Adenoviral vector encoding human atonal homolog-1 (HATH1)
US9951351B2 (en) 2014-10-09 2018-04-24 Genvec, Inc. Adenoviral vector encoding human atonal homolog-1 (HATH1)
WO2016131945A1 (en) 2015-02-20 2016-08-25 Transgene Sa Combination product with autophagy modulator
WO2017191147A1 (en) 2016-05-04 2017-11-09 Transgene Sa Combination therapy with cpg tlr9 ligand
WO2018064523A1 (en) 2016-09-30 2018-04-05 Genvec, Inc. Adenovectors for delivery of therapeutic genetic material into t cells
WO2018069316A2 (en) 2016-10-10 2018-04-19 Transgene Sa Immunotherapeutic product and mdsc modulator combination therapy
WO2018129268A1 (en) 2017-01-07 2018-07-12 Selecta Biosciences, Inc. Patterned dosing of immunosuppressants coupled to synthetic nanocarriers
US11299713B2 (en) 2017-04-21 2022-04-12 Geneuin-Tech Co., Ltd. Cell line for producing adenovirus and method of preparing the same
WO2019075360A1 (en) 2017-10-13 2019-04-18 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector igm responses
US11773142B2 (en) 2017-12-11 2023-10-03 Beth Israel Deaconess Medical Center, Inc. Recombinant adenoviruses and uses thereof
US11608362B2 (en) 2018-03-06 2023-03-21 Precigen, Inc. Hepatitis B vaccines and uses of the same
WO2020223205A1 (en) 2019-04-28 2020-11-05 Selecta Biosciences, Inc. Methods for treatment of subjects with preexisting immunity to viral transfer vectors
WO2020243261A1 (en) 2019-05-28 2020-12-03 Selecta Biosciences, Inc. Methods and compositions for attenuated anti-viral transfer vector immune response
WO2022074464A2 (en) 2020-03-05 2022-04-14 Neotx Therapeutics Ltd. Methods and compositions for treating cancer with immune cells
WO2021215919A1 (en) 2020-04-21 2021-10-28 Polpharma Biologics Utrecht B.V. Humanized anti-human cd89 antibodies and uses thereof
WO2022013221A1 (en) 2020-07-13 2022-01-20 Transgene Treatment of immune depression
KR20230038496A (en) 2020-07-13 2023-03-20 트랜스진 treatment of immunosuppression
WO2023017494A1 (en) 2021-08-13 2023-02-16 Triovance Holding Llc A skin substitute composition and methods of producing and using the same
WO2023064367A1 (en) 2021-10-12 2023-04-20 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector igm responses
WO2023172624A1 (en) 2022-03-09 2023-09-14 Selecta Biosciences, Inc. Immunosuppressants in combination with anti-igm agents and related dosing
WO2023213764A1 (en) 2022-05-02 2023-11-09 Transgene Fusion polypeptide comprising an anti-pd-l1 sdab and a member of the tnfsf

Also Published As

Publication number Publication date
EP1445322B2 (en) 2012-06-06
KR100470180B1 (en) 2005-02-04
US20020164802A1 (en) 2002-11-07
DK1445322T3 (en) 2010-01-04
US20020151032A1 (en) 2002-10-17
US20090023196A1 (en) 2009-01-22
SI1445322T1 (en) 2010-01-29
DK0833934T3 (en) 2005-02-07
US6395519B1 (en) 2002-05-28
DE69633565D1 (en) 2004-11-11
JP2006345872A (en) 2006-12-28
DK0833934T4 (en) 2012-11-19
ATE445705T1 (en) 2009-10-15
DE69638058D1 (en) 2009-11-26
AU6018296A (en) 1997-01-15
AU731767B2 (en) 2001-04-05
SI0833934T1 (en) 2005-02-28
CA2222140A1 (en) 1997-01-03
KR19990022941A (en) 1999-03-25
EP0833934A1 (en) 1998-04-08
US6238893B1 (en) 2001-05-29
US8236293B2 (en) 2012-08-07
EP0833934B2 (en) 2012-08-15
JPH11507552A (en) 1999-07-06
DK1445322T4 (en) 2012-09-03
ATE278794T1 (en) 2004-10-15
EP1445322B1 (en) 2009-10-14
EP0833934B1 (en) 2004-10-06
CA2222140C (en) 2010-11-23
US6306652B1 (en) 2001-10-23
SI1445322T2 (en) 2012-10-30
KR100449181B1 (en) 2005-01-24
ES2231813T5 (en) 2012-12-17
PT1445322E (en) 2010-01-19
ES2333425T5 (en) 2012-08-28
ES2333425T3 (en) 2010-02-22
ES2231813T3 (en) 2005-05-16
DE69633565T3 (en) 2013-01-17
US6602706B1 (en) 2003-08-05
DE69633565T2 (en) 2005-12-22
PT833934E (en) 2005-02-28
EP1445322A1 (en) 2004-08-11
US6033908A (en) 2000-03-07
US5994128A (en) 1999-11-30
JP4051416B2 (en) 2008-02-27
IL160406A (en) 2008-03-20
KR20040065317A (en) 2004-07-21
SI0833934T2 (en) 2013-04-30
IL122614A0 (en) 1998-08-16
US20050221492A1 (en) 2005-10-06
IL160406A0 (en) 2004-07-25
HK1064406A1 (en) 2005-01-28

Similar Documents

Publication Publication Date Title
EP0833934B1 (en) Packaging systems for human recombinant adenovirus to be used in gene therapy
US6670188B1 (en) Packaging systems for human recombinant adenovirus to be used in gene therapy
CA2378061A1 (en) Packaging systems for human recombinant adenovirus to be used in gene therapy
US20050158278A1 (en) Means and methods for the production of adenovirus vectors
US20080138901A1 (en) Packaging systems for human recombinant adenovirus to be used in gene therapy
AU766771B2 (en) Packaging systems for human recombinant adenoviruses to be used in gene therapy
AU2003227281B2 (en) Packaging systems for human recombinant adenoviruses to be used in gene therapy
MXPA99011998A (en) Generation of packaging system for human recombinant adenoviral vectors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IL IS JP KE KG KP KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 08793170

Country of ref document: US

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2222140

Country of ref document: CA

Kind code of ref document: A

Ref document number: 1997 502948

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1019970709411

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1996917735

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1996917735

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1019970709411

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 160406

Country of ref document: IL

WWR Wipo information: refused in national office

Ref document number: 1019970709411

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 1996917735

Country of ref document: EP

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)