US20050169891A1 - Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells - Google Patents

Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells Download PDF

Info

Publication number
US20050169891A1
US20050169891A1 US11/018,669 US1866904A US2005169891A1 US 20050169891 A1 US20050169891 A1 US 20050169891A1 US 1866904 A US1866904 A US 1866904A US 2005169891 A1 US2005169891 A1 US 2005169891A1
Authority
US
United States
Prior art keywords
adenovirus
cells
subgroup
protein
recombinant
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/018,669
Inventor
Ronald Vogels
Menzo Havenga
Abraham Bout
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Vaccines and Prevention BV
Original Assignee
Crucell Holand BV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/348,354 external-priority patent/US20030017138A1/en
Application filed by Crucell Holand BV filed Critical Crucell Holand BV
Priority to US11/018,669 priority Critical patent/US20050169891A1/en
Publication of US20050169891A1 publication Critical patent/US20050169891A1/en
Assigned to CRUCELL HOLLAND B.V. reassignment CRUCELL HOLLAND B.V. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BOUT, ABRAHAM, HAVENGA, MENZO J.E., VOGELS, RONALD
Priority to US12/455,086 priority patent/US7968087B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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
    • C12N2710/10322New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10345Special targeting system for viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/60Vectors comprising as targeting moiety peptide derived from defined protein from viruses
    • C12N2810/6009Vectors comprising as targeting moiety peptide derived from defined protein from viruses dsDNA viruses
    • C12N2810/6018Adenoviridae

Definitions

  • the invention relates generally to biotechnology and, more particularly, to the field of molecular genetics and medicine.
  • the present invention relates to the field of gene therapy and, more particularly, to gene therapy using adenoviruses.
  • genetic information is usually delivered to a host cell in order to either correct (supplement) a genetic deficiency in the host cell, or to inhibit an undesired function in the host cell, or to eliminate the host cell.
  • the genetic information can also be intended to provide the host cell with a desired function, e.g., to supply a secreted protein to treat other cells of the host, etc.
  • adenovirus vectors for the delivery of nucleic acid to target cells in gene therapy protocols.
  • some characteristics of the current vectors limit their use in specific applications. For instance, endothelial cells and smooth muscle cells are not easily transduced by the current generation of adenovirus vectors.
  • these types of cells should be genetically modified.
  • even the very good in vivo delivery capacity of adenovirus vectors is not sufficient and higher transfer efficiencies are required. This is the case, for instance, when most cells of a target tissue need to be transduced.
  • the present invention was made in the course of the manipulation of adenovirus vectors. In the following section, therefore, a brief introduction to adenoviruses is given.
  • Adenoviruses contain a linear double-stranded DNA molecule of approximately 36000 base pairs.
  • the DNA molecule contains identical Inverted Terminal Repeats (ITR) of approximately 90-140 base pairs with the exact length depending on the serotype.
  • ITR Inverted Terminal Repeats
  • the viral origins of replication are within the ITRs exactly at the genome ends.
  • the transcription units are divided into early and late regions. Shortly after infection, the E1A and E1B proteins are expressed and function in transactivation of cellular and adenoviral genes.
  • the early regions E2A and E2B encode proteins (DNA binding protein, pre-terminal protein, and polymerase) required for the replication of the adenoviral genome (reviewed in van der Vliet, 1995).
  • the early region E4 encodes several proteins with pleiotropic functions, e.g., transactivation of the E2 early promoter, facilitating transport and accumulation of viral mRNAs in the late phase of infection and increasing nuclear stability of major late pre-mRNAs (reviewed in Leppard, 1997).
  • the early region 3 encodes proteins that are involved in modulation of the immune response of the host (Wold et al., 1995).
  • the late region is transcribed from one single promoter (major late promoter) and is activated at the onset of DNA replication.
  • RNA species coding for core proteins, capsid proteins (Penton, hexon, fiber and associated proteins), viral protease and proteins necessary for the assembly of the capsid and shut-down of host protein translation (Imperiale, M. J., Akusjnarvi, G. and Leppard, K. N. (1995). Post-transcriptional control of adenovirus gene expression. In: The molecular repertoire of adenoviruses I. P139-171. W. Doerfler and P. Bohm (eds), springer-Verlag Berlin Heidelberg).
  • the interaction of the virus with the host cell has mainly been investigated with the serotype C viruses Ad2 and Ad5. Binding occurs via interaction of the knob region of the protruding fiber with a cellular receptor.
  • the receptor for Ad2 and Ad5 and probably more adenoviruses is known as the “Coxsackievirus and Adenovirus Receptor” or CAR protein (Bergelson et al., 1997). Internalization is mediated through interaction of the RGD sequence present in the penton base with cellular integrins (Wickham et al., 1993). This may not be true for all serotypes, for example, serotypes 40 and 41 do not contain a RGD sequence in their penton base sequence (Kidd et al., 1993).
  • the Fiber Protein The Fiber Protein:
  • the initial step for successful infection is binding of adenovirus to its target cell, a process mediated through fiber protein.
  • the fiber protein has a trimeric structure (Stouten et al., 1992) with different lengths depending on the virus serotype (Signas et al., 1985; Kidd et al., 1993). Different serotypes have polypeptides with structurally similar N and C termini, but different middle stem regions. The first 30 amino acids at the N terminus are involved in anchoring of the fiber to the penton base (Chroboczek et al., 1995), especially the conserved FNPVYP (SEQ ID NO:_______) region in the tail (Arnberg et al., 1997).
  • the C-terminus is responsible for initial interaction with the cellular adenovirus receptor. After this initial binding, secondary binding between the capsid penton base and cell-surface integrins leads to internalization of viral particles in coated pits and endocytosis (Morgan et al., 1969; Svensson and Persson, 1984; Varga et al., 1991; Greber et al., 1993; Wickham et al., 1993). Integrins are ⁇ -heterodimers of which at least 14 ⁇ -subunits and 8 ⁇ -subunits have been identified (Hynes, 1992). The array of integrins expressed in cells is complex and will vary between cell types and cellular environment. Although the knob contains some conserved regions, between serotypes, knob proteins show a high degree of variability, indicating that different adenovirus receptors exist.
  • a serotype is defined on the basis of its immunological distinctiveness as determined by quantitative neutralization with animal antiserum (horse, rabbit).
  • serotype is assumed if A) the hemagglutinins are unrelated, as shown by lack of cross-reaction on hemagglutination-inhibition, or B) substantial biophysical/biochemical differences in DNA exist (Francki et al., 1991).
  • the serotypes identified last (42-49) were isolated for the first time from HIV infected patients (Hierholzer et al., 1988; Schnurr et al., 1993). For reasons not well understood, most of such immuno-compromised patients shed adenoviruses that were never isolated from immuno-competent individuals (Hierholzer et al., 1988, 1992; Khoo et al., 1995).
  • adenoviruses in subgroup C such as Ad2 and Ad5 bind to different receptors as compared to adenoviruses from subgroup B such as Ad3 and Ad7 (Defer et al., 1990; Gall et al., 1996).
  • receptor specificity could be altered by exchanging the Ad3 knob protein with the Ad 5 knob protein, and vice versa (Krasnykh et al., 1996; Stevenson et al., 1995, 1997).
  • Serotypes 2, 4, 5 and 7 all have a natural affiliation towards lung epithelia and other respiratory tissues.
  • serotypes 40 and 41 have a natural affiliation towards the gastrointestinal tract. These serotypes differ in at least capsid proteins (penton-base, hexon), proteins responsible for cell binding (fiber protein), and proteins involved in adenovirus replication. It is unknown to what extent the capsid proteins determine the differences in tropism found between the serotypes. It may very well be that post-infection mechanisms determine cell type specificity of adenoviruses.
  • adenoviruses from serotypes A (Ad12 and Ad31), C (Ad2 and Ad5), D (Ad9 and Ad15), E (Ad4) and F (Ad41) all are able to bind labeled, soluble CAR (sCAR) protein when immobilized on nitrocellulose. Furthermore, binding of adenoviruses from these serotypes to Ramos cells, that express high levels of CAR but lack integrins (Roelvink et al., 1996), could be efficiently blocked by addition of sCAR to viruses prior to infection (Roelvink et al., 1998).
  • subgroup D serotypes have relatively short fiber shafts compared to subgroup A and C viruses. It has been postulated that the tropism of subgroup D viruses is to a large extent determined by the penton base binding to integrins (Roelvink et al., 1996; Roelvink et al., 1998).
  • adenoviral gene delivery vectors currently used in gene therapy are derived from the serotype C adenoviruses Ad2 or Ad5.
  • the vectors have a deletion in the E1 region, where novel genetic information can be introduced.
  • the E1 deletion renders the recombinant virus replication defective. It has been demonstrated extensively that recombinant adenovirus, in particular serotype 5, is suitable for efficient transfer of genes in vivo to the liver, the airway epithelium and solid tumors in animal models and human xenografts in immuno-deficient mice (Bout 1996, 1997; Blaese et al., 1995).
  • Gene transfer vectors derived from adenoviruses have a number of features that make them particularly useful for gene transfer:
  • the present invention provides gene therapy methods, compounds and medicines.
  • the present invention is particularly useful in gene therapy applications where endothelial cells and/or smooth muscle cells form the target cell type.
  • the present invention relates to gene delivery vehicles provided with a tissue tropism for at least endothelial cells and/or smooth muscle cells.
  • the present invention further relates to gene delivery vehicles having been deprived of a tissue tropism for liver cells.
  • Table I Oligonucleotides and degenerate oligonucleotides used for the amplification of DNA encoding fiber proteins derived from alternative adenovirus serotypes. (Bold letters represent NdeI restriction site (A-E), NsiI restriction site (1-6, 8), or PacI restriction site (7).
  • Table III Expression of CAR and integrins on the cell surface of endothelial cells and smooth muscle cells.
  • 70% Cells harvested for FACS analysis at a cell density of 70% confluency.
  • 100% Cells harvested for FACS analysis at a cell density of 100% confluency.
  • PER.C6 cells were taken as a control for antibody staining. Values represent percentages of cells that express CAR or either one of the integrins at levels above background. As background control, HUVECs or HUVsmc were incubated only with the secondary, rat-anti-mouse IgG1 labeled antibody.
  • Table IV Determination of transgene expression (luciferase activity) per ⁇ g of total cellular protein, after infection of A549 cells.
  • FIG. 1 Schematic drawing of the pBr/Ad.Bam-rITR construct.
  • FIG. 2 Schematic drawing of the strategy used to delete the fiber gene from the pBr/Ad.Bam-rITR construct.
  • FIG. 3 Schematic drawing of construct pBr/Ad.BamR ⁇ fib (ECACC deposit number 01121708).
  • FIG. 4 Sequences of the chimeric fibers Ad5 (SEQ ID NO:16) Ad5/12 (SEQ ID NO:17), Ad5/16 (SEQ ID NO:18), Ad5/28 (SEQ ID NO:19), and Ad5/40-L (SEQ ID NO:20).
  • FIG. 5 Schematic drawing of the construct pCArchitectl.-Luc.
  • FIG. 6 Schematic drawing of the method to generate chimeric adenoviruses using three overlapping fragments. Early (E) and late regions (L) are indicated. L5 is the fiber coding sequence.
  • FIG. 7 A) Infection of HUVEC cells using different amounts of virus particles per cell and different fiber chimeric adenoviruses.
  • RLU relative light units
  • HUVsmc were infected with 312 (light grey bar), 625 (grey bar), 1250 (black bar), 2500 (dark grey bar), 5000 (light grey bar), or 10,000 (white bar) virus particles per cell of either the fiber 11, 16, 35, or 51 chimeric virus. Luciferase transgene expression expressed as RLU per microgram protein was measured 48 hours after virus exposure.
  • FIG. 9 Sequences including the gene encoding adenovirus 16 fiber protein as published in Genbank (SEQ ID NO:21) and sequence including a gene encoding a fiber from an adenovirus 16 variant as isolated in the present invention, wherein the sequences of the fiber protein are from the NdeI-site (SEQ ID NO:22).
  • FIG. 9A is a nucleotide sequence comparison (SEQ ID NOS:21 (upper stand) and 22 (lower strand)).
  • FIG. 9B is an amino-acid comparison (SEQ ID NOS:23 (upper strand) and 24 (lower strand)).
  • the current invention provides materials and methods to overcome the limitations of adenoviral vectors mentioned above.
  • the invention provides new adenoviruses, derived in whole or in part from serotypes different from Ad5. Specific genes of serotypes with preferred characteristics may be combined in a chimeric vector to give rise to a vector that is better suited for specific applications.
  • Preferred characteristics include, but are not limited to, improved infection of a specific target cell, reduced infection of non-target cells, improved stability of the virus, reduced uptake in antigen presenting cells (APC), or increased uptake in APC, reduced toxicity to target cells, reduced neutralization in humans or animals, reduced or increased CTL response in humans or animals, better and/or prolonged transgene expression, increased penetration capacity in tissues, improved yields in packaging cell lines, etc.
  • APC antigen presenting cells
  • One aspect of the present invention facilitates the combination of the low immunogenicity of some adenoviruses with the characteristics of other adenoviruses that allow efficient gene therapy.
  • Such characteristics may be a high specificity for certain host cells, a good replication machinery for certain cells, a high rate of infection in certain host cells, low infection efficiency in non-target cells, high or low efficiency of APC infection, etc.
  • the invention thus may provide chimeric adenoviruses having the useful properties of at least two adenoviruses of different serotypes.
  • the present invention provides adenovirus vectors that can be used as cassettes to insert different adenoviral genes from different adenoviral serotypes at the required sites.
  • a vector capable of producing a chimeric adenovirus whereby of course also a gene of interest can be inserted (for instance at the site of E1 of the original adenovirus).
  • the chimeric adenovirus to be produced can be adapted to the requirements and needs of certain hosts in need of gene therapy for certain disorders.
  • a packaging cell will generally be needed in order to produce sufficient amount of safe chimeric adenoviruses.
  • the present invention provides adenoviral vectors comprising at least a fragment of a fiber protein of an adenovirus from subgroup B.
  • This fiber protein may be the native fiber protein of the adenoviral vector or may be derived from a serotype different from the serotype the adenoviral vector is based on.
  • the adenoviral vector according to the invention is a chimeric adenovirus displaying at least a fragment of the fiber protein derived from subgroup B adenovirus, wherein the fragment comprises at least the receptor binding sequence.
  • a virus will be produced using a vector (typically a plasmid, a cosmid or baculovirus vector).
  • a preferred vector is a vector that can be used to make a chimeric recombinant virus specifically adapted to the host to be treated and the disorder to be treated.
  • the present invention also provides a chimeric adenovirus based on adenovirus type 5, but having at least a fragment of the fiber sequence from adenovirus type 16, wherein the fragment of the fiber of Ad16 comprises the fragment of the fiber protein that is involved in binding a host cell.
  • the present invention also provides chimeric adenoviral vectors that show improved infection as compared to adenoviruses from other subgroups in specific host cells, for example, but not limited to, endothelial cells and smooth muscle cells of human or animal origin.
  • An important feature of the present invention is the means to produce the chimeric virus. Typically, one does not want an adenovirus batch to be administered to the host cell, which contains replication competent adenovirus.
  • a cell is usually called a “packaging cell.”
  • the invention thus also provides a packaging cell for producing a chimeric adenovirus according to the invention, comprising, in trans, all elements necessary for adenovirus production not present on the adenoviral vector according to the invention.
  • vector and packaging cell are adapted to one another in that they have all the necessary elements, but that they do not have overlapping elements which lead to replication competent virus by recombination.
  • the invention also provides a kit of parts comprising a packaging cell according to the invention and a recombinant vector according to the invention wherein there is essentially no sequence overlap leading to recombination resulting in the production of replication competent adenovirus between the cell and the vector.
  • adenoviral vector according to the invention may be replication competent or capable of replicating under certain conditions, for example, in specific cell types like tumor cells or tumor endothelial cells.
  • the chimeric adenoviral vector according to the invention may originate from at least two different serotypes. This may provide the vector with preferred characteristics such as improved infection of target cells and/or less infection of non-target cells, improved stability of the virus, reduced immunogenicity in humans or animals (e.g., reduced uptake in APC, reduced neutralization in the host and/or reduced cytotoxic T-lymphocyte (CTL) response), increased penetration of tissue, better longevity of transgene expression, etc.
  • CTL cytotoxic T-lymphocyte
  • capsid genes e.g., penton and/or hexon genes from less immunogenic serotypes as defined by the absence or the presence of low amounts of neutralizing antibodies in the vast majority of hosts.
  • fiber and/or penton sequences from serotypes that show improved binding and internalization in the target cells.
  • a vector deleted of all adenoviral genes is also preferred.
  • the promoter driving the gene of interest to be expressed in the target cells is a cell type specific promoter.
  • a library of adenoviral genes be provided wherein the genes to be exchanged are located on plasmid- or cosmid-based adenoviral constructs wherein the genes or the sequences to be exchanged are flanked by restriction sites.
  • the preferred genes or sequences can be selected from the library and inserted in the adenoviral constructs that are used to generate the viruses.
  • such a method comprises a number of restriction and ligation steps and transfection of a packaging cell.
  • the adenoviral vector can be transfected in one piece, or as two or more overlapping fragments, wherein viruses are generated by homologous recombination.
  • the adenoviral vector may be built up from two or more overlapping sequences for insertion or replacements of a gene of interest in, for example, the E1 region, for insertion or replacements in penton and/or hexon sequences, and for insertions or replacements into fiber sequences.
  • the invention provides a method for producing chimeric adenoviruses having one or more desired properties like a desired host range and diminished antigenicity, comprising providing one or more vectors according to the invention having the desired insertion sites, inserting into these vectors at least a functional part of a fiber protein derived from an adenovirus serotype having the desired host range and/or inserting a functional part of a capsid protein derived from an adenovirus serotype having a relatively low antigenicity and transfecting these vectors in a packaging cell according to the invention and allowing for production of chimeric viral particles.
  • other combinations of other viral genes originating from different serotypes can also be inserted as disclosed herein before.
  • Chimeric viruses having only one non-native sequence in addition to an insertion or replacement of a gene of interest in the E1 region are also within the scope of the invention.
  • An immunogenic response to adenovirus that typically occurs is the production of neutralizing antibodies by the host. This response is typically a reason for selecting a capsid protein like penton, hexon and/or fiber of a less immunogenic serotype.
  • the invention also provides a chimeric adenovirus wherein the hexon, penton, fiber and/or other capsid proteins are chimeric proteins originating from different adenovirus serotypes.
  • capsid (protein) genes etc. carrying non-adenoviral sequences or mutations, such as point mutations, deletions, insertions, etc., which can be easily screened for preferred characteristics such as temperature stability, assembly, anchoring, redirected infection, altered immune response etc.
  • capsid (protein) genes etc. carrying non-adenoviral sequences or mutations, such as point mutations, deletions, insertions, etc., which can be easily screened for preferred characteristics such as temperature stability, assembly, anchoring, redirected infection, altered immune response etc.
  • other chimeric combinations can also be produced and are within the scope of the present invention.
  • mice and rats that upon in vivo systemic delivery of recombinant adenovirus of commonly used serotypes for gene therapy purposes more than 90% of the virus is trapped in the liver, (Herz et al., 1993; Kass-Eisler et al., 1994; Huard et al., 1995). It is also known that human hepatocytes are efficiently transduced by adenovirus serotype 5 vectors (Castell, J. V., Hernandez, D. Gomez-Foix, A. M., Guillen, I, Donato, T. and Gomez-Lechon, M. J. (1997).
  • Adenovirus-mediated gene transfer into human hepatocytes analysis of the biochemical functionality of transduced cells. Gene Ther. 4 (5), p 455-464).
  • Ad2 or Ad5 based vectors are seriously hampered by the efficient uptake of the viruses in the liver leading to unwanted toxicity and less virus being available for transduction of the target cells. Therefore, alteration of the adenovirus serotype 5 host cell range to be able to target other organs in vivo is a major interest of the invention.
  • Wickham et al. have altered the RGD (Arg, Gly, Asp) motif in the penton base which is believed to be responsible for the ⁇ 3 and ⁇ 5 integrin binding to the penton base. They have replaced this RGD motif by another peptide motif which is specific for the ⁇ 4 ⁇ 1 receptor. In this way, targeting the adenovirus to a specific target cell could be accomplished (Wickham et al., 1995). Krasnykh et al. (1998) have made use of the HI loop available in the knob.
  • This loop is, based on X-ray crystallography, located on the outside of the knob trimeric structure and therefore is thought not to contribute to the intramolecular interactions in the knob. Insertion of a FLAG coding sequence into the HI loop resulted in fiber proteins that were able to trimerize and it was further shown that viruses containing the FLAG sequence in the knob region could be made. Although interactions of the FLAG-containing knob with CAR are not changed, insertion of ligands in the HI loop may lead to retargeting of infection. Although successful introduction of changes in the adenovirus serotype 5 fiber and penton-base have been reported, the complex structure of knob and the limited knowledge of the precise amino acids interacting with CAR render such targeting approaches laborious and difficult. The use of antibodies binding to CAR and to a specific cellular receptor has also been described (Wickham et al., 1996; Rogers et al., 1997). This approach is however limited by the availability of specific antibody and by the complexity of the gene therapy product.
  • the present invention provides methods for the generation of chimeric capsids which can be targeted to specific cell types in vitro as well as in vivo, and thus have an altered tropism for certain cell types.
  • the present invention further provides methods and means by which an adenovirus or an adenovirus capsid can be used as a protein or nucleic acid delivery vehicle to a specific cell type or tissue.
  • chimeric adenoviruses based on adenovirus serotype 5 with modified late genes is described.
  • three plasmids which together contain the complete adenovirus serotype 5 genome, were constructed. From one of these plasmids part of the DNA encoding the adenovirus serotype 5 fiber protein was removed and replaced by linker DNA sequences that facilitate easy cloning. This plasmid subsequently served as template for the insertion of DNA encoding fiber protein derived from different adenovirus serotypes.
  • the DNA sequences derived from the different serotypes were obtained using the polymerase chain reaction technique in combination with (degenerate) oligonucleotides.
  • any gene of interest can be cloned.
  • a single transfection procedure of the three plasmids together results in the formation of a recombinant chimeric adenovirus.
  • cloning of the sequences obtained from the library of genes can be such that the chimeric adenoviral vector is built up from one or two fragments.
  • one construct contains at least the left ITR and sequences necessary for packaging of the virus, an expression cassette for the gene of interest and sequences overlapping with the second construct comprising all sequences necessary for replication and virus formation not present in the packaging cell as well as the non-native sequences providing the preferred characteristics.
  • This new technology of libraries consisting of chimeric adenoviruses thus allows for a rapid screening improved recombinant adenoviral vectors for in vitro and in vivo gene therapy purposes.
  • adenovirus type 5 for in vivo gene therapy is limited by the apparent inability to infect certain cell types, e.g., human endothelial cells and smooth muscle cells and the preference of infection of certain organs, e.g., liver and spleen. Specifically, this has consequences for treatment of cardiovascular diseases like restenosis and pulmonary hypertension.
  • Adenovirus-mediated delivery of human ceNOS (constitutive endothelial nitric oxide synthase) has been proposed as treatment for restenosis after percutaneous transluminal coronary angioplasty (PTCA).
  • Restenosis is characterized by progressive arterial remodeling, extracellular matrix formation and intimal hyperplasia at the site of angioplasty (Schwartz et al., 1993; Carter et al., 1994; Shi et al., 1996).
  • NO is one of the vasoactive factors shown to be lost after PTCA-induced injury to the endothelial barrier (Lloyd Jones and Bloch, 1996).
  • restoration of NO levels after balloon-induced injury by means of adenoviral delivery of ceNOS may prevent restenosis (Varenne et al., 1998).
  • viruses or chimeric viruses according to the invention are superior to Ad2 or Ad5 based viruses, given as non-limiting examples, are production of proteins by endothelial cells that are secreted into the blood, treatment of hypertension, preventive treatment of stenosis during vein grafting, angiogenesis, heart failure, renal hypertension and others.
  • this invention includes adenoviral vectors that are, amongst others, especially suited for gene delivery to endothelial cells and smooth muscle cells important for treatment of cardiovascular disorders.
  • the adenoviral vectors preferably are derived from subgroup B adenoviruses or contain at least a functional part of the fiber protein from an adenovirus from subgroup B comprising at least the cell-binding moiety of the fiber protein.
  • the adenoviral vectors are chimeric vectors based on adenovirus type 5 and contain at least a functional part of the fiber protein from adenovirus type 16.
  • this invention provides adenoviral vectors or chimeric adenoviral vectors that escape the liver following systemic administration.
  • these adenoviral vectors are derived from subgroup A, B, D, or F in particular serotypes 12, 16, 28 and 40 or contain at least the cell-binding moiety of the fiber protein derived from the adenoviruses.
  • the adenoviral vectors may be derived from the serotype having the desired properties or that the adenoviral vector is based on an adenovirus from one serotype and contains the sequences comprising the desired functions of another serotype, these sequences replacing the native sequences in the serotype.
  • this invention describes chimeric adenoviruses and methods to generate these viruses that have an altered tropism different from that of adenovirus serotype 5. For example, viruses based on adenovirus serotype 5 but displaying any adenovirus fiber existing in nature. This chimeric adenovirus serotype 5 is able to infect certain cell types more efficiently, or less efficiently in vitro and in vivo than the adenovirus serotype 5.
  • Such cells include, but are not limited to, endothelial cells, smooth muscle cells, dendritic cells, neuronal cells, glial cells, synovical cells, lung epithelia cells, hemopoietic stem cells, monocytic/macrophage cells, tumor cells, skeletal muscle cells, mesothelial cells, synoviocytes, etc.
  • the invention describes the construction and use of libraries consisting of distinct parts of adenovirus serotype 5 in which one or more genes or sequences have been replaced with DNA derived from alternative human or animal serotypes.
  • This set of constructs in total encompassing the complete adenovirus genome, allows for the construction of unique chimeric adenoviruses customized for a certain disease, group of patients or even a single individual.
  • the chimeric adenoviruses may, or may not, contain deletions in the E1 region and insertions of heterologous genes linked either or not to a promoter. Furthermore, chimeric adenoviruses may, or may not, contain deletions in the E3 region and insertions of heterologous genes linked to a promoter. Furthermore, chimeric adenoviruses may, or may not, contain deletions in the E2 and/or E4 region and insertions of heterologous genes linked to a promoter. In the latter case, E2 and/or E4 complementing cell lines are used to generate recombinant adenoviruses.
  • any gene in the genome of the viral vector can be taken out and supplied in trans.
  • chimeric viruses do not contain any adenoviral genes in their genome and are by definition situation minimal adenoviral vectors. In this case all adenoviral functions are supplied in trans using stable cell lines and/or transient expression of these genes.
  • a method for producing minimal adenoviral vectors is described in PCT International Publication WO97/00326 and is incorporated by reference herein.
  • Ad/AAV chimeric molecules are packaged into the adenovirus capsids of the invention.
  • a method for producing Ad/AAV chimeric vectors is described in European Patent Office publication EP 1 042 494 and is incorporated by reference herein.
  • any nucleic acid may be provided with the adenovirus capsids of the invention.
  • the invention provides a gene delivery vehicle having been provided with at least a tissue tropism for smooth muscle cells and/or endothelial cells. In another embodiment, the invention provides a gene delivery vehicle deprived of a tissue tropism for at least liver cells. Preferably, the gene delivery vehicle is provided with a tissue tropism for at least smooth muscle cells and/or endothelial cells and deprived of a tissue tropism for at least liver cells. In a preferred embodiment, the gene delivery vehicle is provided with a tissue tropism for at least smooth muscle cells and/or endothelial cells and/or deprived of a tissue tropism for at least liver cells using a fiber protein derived from a subgroup B adenovirus, preferably of adenovirus 16.
  • the gene delivery vehicle comprises a virus capsid.
  • this virus capsid comprises a virus capsid derived in whole or in part from an adenovirus of subgroup B, preferably from adenovirus 16, or it comprises proteins, or parts thereof, from an adenovirus of subgroup B, preferably of adenovirus 16.
  • the virus capsid comprises proteins, or fragments thereof, from at least two different viruses, preferably adenoviruses.
  • at least one of the viruses is an adenovirus of subgroup B, preferably adenovirus 16.
  • the gene delivery vehicle comprises an adenovirus fiber protein or fragments thereof.
  • the fiber protein is preferably derived from an adenovirus of subgroup B, preferably adenovirus 16.
  • the gene delivery vehicle may further comprise other fiber proteins, or fragments thereof, from other adenoviruses.
  • the gene delivery vehicle may or may not comprise other adenovirus proteins.
  • Nucleic acid may be linked directly to fiber proteins, or fragments thereof, but may also be linked indirectly.
  • indirect linkages include, but are not limited to, packaging of nucleic acid into adenovirus capsids or packaging of nucleic acid into liposomes, wherein a fiber protein, or a fragment thereof, is incorporated into an adenovirus capsid or linked to a liposome.
  • Direct linkage of nucleic acid to a fiber protein, or fragment thereof may be performed when the fiber protein, or a fragment thereof, is not part of a complex or when the fiber protein or a fragment thereof, is part of a complex, such as an adenovirus capsid.
  • a gene delivery vehicle comprising an adenovirus fiber protein wherein the fiber protein comprises a tissue-determining fragment of an adenovirus of subgroup B adenovirus preferably of adenovirus 16.
  • Adenovirus fiber protein comprises three functional domains. One domain, the base, is responsible for anchoring the fiber to a penton base of the adenovirus capsid. Another domain, the knob, is responsible for receptor recognition whereas the shaft domain functions as a spacer separating the base from the knob. The different domains may also have other functions. For instance, the shaft is presumably also involved in target cell specificity. Each of the domains mentioned above may be used to define a fragment of a fiber. However, fragments may also be identified in another way. For instance, the knob domain comprises a receptor binding fragment and a shaft binding fragment. The base domain comprises of a penton base binding fragment and a shaft binding fragment. Moreover, the shaft comprises repeated stretches of amino acids. Each of these repeated stretches may be a fragment.
  • Fiber proteins possess tissue tropism-determining properties. The most well described fragment of the fiber protein involved in tissue tropism is the knob domain. However, the shaft domain of the fiber protein also possesses tissue tropism-determining properties. However, not all of the tissue tropism-determining properties of an adenovirus capsid are incorporated into a fiber protein.
  • tissue tropism-determining fragment of a fiber protein may be a single fragment of a fiber protein or a combination of fragments of at least one fiber protein, wherein the tissue tropism-determining fragment, either alone or in combination with a virus capsid, determines the efficiency with which a gene delivery vehicle can transduce a given cell or cell type, preferably but not necessarily in a positive way.
  • tissue tropism for liver cells is meant a tissue tropism for cells residing in the liver, preferably liver parenchyma cells.
  • a tissue tropism for a certain tissue may be provided by increasing the efficiency with which cells of the tissue are transduced, alternatively, a tissue tropism for a certain tissue may be provided by decreasing the efficiency with which other cells than the cells of the tissue are transduced.
  • a fiber protein derived from a subgroup B adenovirus is combined with the non-fiber capsid proteins from an adenovirus of subgroup C, preferably of adenovirus 5.
  • a gene delivery vehicle comprising a nucleic acid derived from an adenovirus.
  • the adenovirus nucleic acid comprises at least one nucleic acid sequence encoding a fiber protein comprising at least a tissue tropism-determining fragment of a subgroup B adenovirus fiber protein, preferably of adenovirus 16.
  • the adenovirus comprises nucleic acid from at least two different adenoviruses.
  • the adenovirus comprises nucleic acid from at least two different adenoviruses wherein at least one nucleic acid sequence encoding a fiber protein comprises at least a tissue tropism-determining fragment of a subgroup B adenovirus fiber protein, preferably of adenovirus 16.
  • the adenovirus nucleic acid is modified such that the capacity of the adenovirus nucleic acid to replicate in a target cell has been reduced or disabled. This may be achieved through, for example, inactivating or deleting genes encoding early region proteins.
  • the adenovirus nucleic acid is modified such that the capacity of a host immune system to mount an immune response against adenovirus proteins encoded by the adenovirus nucleic acid has been reduced or disabled. This may be achieved through deletion of genes encoding proteins of early region 2 and/or early region 4. Alternatively, genes encoding early region 3 proteins may be deleted, or on the contrary, considering the anti-immune system function of some of the proteins encoded by the genes in early region 3, the expression of early region 3 proteins may be enhanced for some purposes. Also, the adenovirus nucleic acid may be altered by a combination of two or more of the specific alterations of the adenovirus nucleic acid mentioned above.
  • adenovirus nucleic acid may also be modified such that the capacity of a host immune system to mount an immune response against adenovirus proteins encoded by the adenovirus nucleic acid has been reduced or disabled, in other ways than mentioned above, for instance through exchanging capsid proteins, or fragments thereof, by capsid proteins, or fragments thereof, from other serotypes for which humans do not have, or have low levels of, neutralizing antibodies.
  • capsid proteins may be exchanged for other capsid proteins, or fragments thereof, for which individuals are not capable of, or have a low capacity of, raising an immune response against.
  • An adenovirus nucleic acid may be altered further or instead of one or more of the alterations mentioned above, by inactivating or deleting genes encoding adenovirus late proteins such as, but not limited to, hexon, penton, fiber and/or protein IX.
  • all genes encoding adenovirus proteins are deleted from the adenovirus nucleic acid, turning the nucleic acid into a minimal adenovirus vector.
  • the adenovirus nucleic acid is an Ad/AAV chimeric vector, wherein at least the integration means of an adeno-associated virus (AAV) is incorporated into the adenovirus nucleic acid.
  • AAV adeno-associated virus
  • a vector or a nucleic acid which may or may not be one and the same according to the invention, further comprises at least one non-adenovirus gene.
  • at least one of the non-adenovirus genes is selected from the group of genes encoding: an apolipoprotein, a ceNOS, a herpes simplex virus thymidine kinase, an interleukin-3, an interleukin-1 ⁇ , an (anti) angiogenesis protein such as angiostatin, an anti-proliferation protein, a vascular endothelial growth factor (VGEF), a basic fibroblast growth factor (bFGF), a hypoxia inducible factor 1 ⁇ (HIF-1 ⁇ ), a PAI-1 and a smooth muscle cell anti-migration protein.
  • VGEF vascular endothelial growth factor
  • bFGF basic fibroblast growth factor
  • HIF-1 ⁇ hypoxia inducible factor 1 ⁇
  • PAI-1 a smooth muscle cell anti-migration protein.
  • the invention provides a cell for the production of a gene delivery vehicle provided with at least a tissue tropism for smooth muscle cells and/or endothelial cells. In another aspect, the invention provides a cell for the production of a gene delivery vehicle deprived of at least a tissue tropism for liver cells. In another aspect, the invention provides a cell for the production of a gene delivery vehicle provided with at least a tissue tropism for smooth muscle cells and/or endothelial cells and deprived of at least a tissue tropism for liver cells. In a preferred embodiment, the cell is an adenovirus packaging cell, wherein an adenovirus nucleic acid is packaged into an adenovirus capsid.
  • an adenovirus packaging cell of the invention all proteins required for the replication and packaging of an adenovirus nucleic acid, except for the proteins encoded by early region 1, are provided by genes incorporated in the adenovirus nucleic acid.
  • the early region 1 encoded proteins in this aspect of the invention may be encoded by genes incorporated into the cells genomic DNA.
  • said cell is PER.C6 (ECACG deposit number 96022940).
  • gene products required for the replication and packaging of adenovirus nucleic acid into adenovirus capsid are not provided by an adenovirus nucleic acid, they are provided by the packaging cell, either by transient transfection, or through stable transformation of said packaging cell.
  • a gene product provided by the packaging cell may also be provided by a gene present on said adenovirus nucleic acid.
  • fiber protein may be provided by the packaging cell, for instance through transient transfection, and may be encoded by the adenovirus nucleic acid. This feature can among others be used to generate adenovirus capsids comprising of fiber proteins from two different viruses.
  • the gene delivery vehicles of the invention are useful for the treatment of cardiovascular disease or disease treatable by nucleic acid delivery to endothelial cells or smooth muscle cells.
  • a non-limiting example of the latter is for instance cancer, where the nucleic acid transferred comprises a gene encoding an anti-angiogenesis protein.
  • the gene delivery vehicles of the invention may be used as a pharmaceutical for the treatment of diseases.
  • gene delivery vehicles of the invention may be used for the preparation of a medicament for the treatment of diseases.
  • the invention provides an adenovirus capsid with or provided with a tissue tropism for smooth muscle cells and/or endothelial cells wherein the capsid preferably comprises proteins from at least two different adenoviruses and wherein at least a tissue tropism-determining fragment of a fiber protein is derived from a subgroup B adenovirus, preferably of adenovirus 16.
  • the invention provides an adenovirus capsid deprived of a tissue tropism for liver cells wherein the capsid preferably comprises proteins from at least two different adenoviruses and wherein at least a tissue tropism-determining fragment of a fiber protein is derived from a subgroup B adenovirus, preferably of adenovirus 16.
  • the invention comprises the use of an adenovirus capsid, for the delivery of nucleic acid to smooth muscle cells and/or endothelial cells. In another embodiment, the invention comprises the use of an adenovirus capsid for preventing the delivery of nucleic acid to liver cells.
  • the adenovirus capsids of the invention may be used for the treatment of cardiovascular disease or a disease treatable by nucleic acid delivery to endothelial cells or smooth muscle cells.
  • An example of the latter is, for instance, cancer where the nucleic acid transferred comprises a gene encoding an anti-angiogenesis protein.
  • adenovirus capsids of the invention may be used as a pharmaceutical for the treatment of diseases.
  • adenovirus capsids of the invention may be used for the preparation of a medicament for the treatment of diseases.
  • construct pBr/Ad.BamR ⁇ fib (ECACC deposit number 01121708, deposited on Dec. 12, 2001 with the Centre for Applied Microbiology and Research Authority (European Collection of Animal Cell Cultures), Porton Down, Salisbury, Wiltshire SP4, OJG, United Kingdom, an International Depository Authority, in accordance with the Budapest Treaty is provided, comprising adenovirus 5 sequences 21562-31094 and 32794-35938.
  • construct pBr/AdBamRfib16 (ECACC deposit number 01121710, deposited on Dec. 12, 2001 with the Centre for Applied Microbiology and Research Authority (European Collection of Animal Cell Cultures), Porton Down, Salisbury, Wiltshire SP4, OJG, United Kingdom, an International Depository Authority, in accordance with the Budapest Treaty is provided, comprising adenovirus 5 sequences 21562-31094 and 32794-3598, further comprising an adenovirus 16 gene encoding fiber protein.
  • construct pBr/AdBamR.pac/fib16 (ECACC deposit number 01121709, deposited on Dec. 12, 2001 with the Centre for Applied Microbiology and Research Authority (European Collection of Animal Cell Cultures), Porton Down, Salisbury, Wiltshire SP4, OJG, United Kingdom, an International Depository Authority, in accordance with the Budapest Treaty, comprising adenovirus 5 sequences 21562-31094 and 32794-35938 is provided, comprising an adenovirus 16 gene encoding fiber protein, and further comprising a unique PacI-site in the proximity of the adenovirus 5 right terminal repeat, in the non-adenovirus sequence backbone of the construct.
  • construct pWE/Ad.AflIIrITRfib16 (ECACC deposit number 01121711, deposited on Dec. 12, 2001 with the Centre for Applied Microbiology and Research Authority (European Collection of Animal Cell Cultures), Porton Down, Salisbury, Wiltshire SP4, OJG, United Kingdom, an International Depository Authority is provided, in accordance with the Budapest Treaty comprising Ad5 sequence 3534-31094 and 32794-35938, further comprising an adenovirus 16 gene encoding fiber protein.
  • construct pWE/Ad.AflIIrITRDE2Afib16 (ECACC deposit number 01121712, deposited on Dec. 12, 2001 with the Centre for Applied Microbiology and Research Authority (European Collection of Animal Cell Cultures), Porton Down, Salisbury, Wiltshire SP4, OJG, United Kingdom, an International Depository Authority, in accordance with the Budapest Treaty, is provided comprising Ad5 sequences 3534-22443 and 24033-31094 and 32794-35938, further comprising an adenovirus 16 gene encoding fiber protein.
  • the constructs are used for the generation of a gene delivery vehicle or an adenovirus capsid with a tissue tropism for smooth muscle cells and/or endothelial cells.
  • the invention provides a library of adenovirus vectors, or gene delivery vehicles which may be one and the same or not, comprising a large selection of non-adenovirus nucleic acids.
  • adenovirus genes encoding capsid proteins are used to generate a library of adenovirus capsids comprising proteins derived from at least two different adenoviruses, the adenoviruses preferably being derived from two different serotypes, wherein preferably one serotype is an adenovirus of subgroup B.
  • a library of adenovirus capsids is generated comprising proteins from at least two different adenoviruses and wherein at least a tissue tropism-determining fragment of fiber protein is derived from an adenovirus of subgroup B, preferably of adenovirus 16.
  • a fiber protein of adenovirus 16 preferably comprises the sequence given in FIG. 9 .
  • analogous sequences may be obtained by using codon degeneracy.
  • amino-acid substitutions or insertions or deletions may be performed as long as the tissue tropism-determining property is not significantly altered.
  • Such amino-acid substitutions may be within the same polarity group or without.
  • the fiber coding sequence of adenovirus serotype 5 is located between nucleotides 31042 and 32787.
  • DNA encoding fiber we started with construct pBr/Ad.Bam-rITR ( FIG. 1 ; ECACC deposit P97082122). From this construct a NdeI site was first removed.
  • pBr322 plasmid DNA was digested with NdeI after which protruding ends were filled using Klenow enzyme. This pBr322 plasmid was then re-ligated, digested with NdeI and transformed into E. coli DH5 ⁇ .
  • the obtained pBr/ ⁇ NdeI plasmid was digested with ScaI and SalI and the resulting 3198 bp vector fragment was ligated to the 15349 bp ScaI-SalI fragment derived from pBr/Ad.Bam-rITR, resulting in plasmid pBr/Ad.Bam-rITR ⁇ NdeI which hence contained a unique NdeI site.
  • a PCR was performed with oligonucleotides “NY-up” and “NY-down” ( FIG. 2 ). During amplification, both a NdeI and a NsiI restriction site were introduced to facilitate cloning of the amplified fiber DNAs.
  • Amplification consisted of 25 cycles of each for 45 seconds at 94° C., 1 minute at 60° C., and 45 seconds at 72° C.
  • the PCR reaction contained 25 pmol of oligonucleotides NY-up or NY-down, 2 mM dNTP, PCR buffer with 1.5 mM MgCl 2 , and 1 unit of Elongase heat stable polymerase (Gibco, The Netherlands).
  • One-tenth of the PCR product was run on an agarose gel which demonstrated that the expected DNA fragment of ⁇ 2200 bp was amplified. This PCR fragment was subsequently purified using Geneclean kit system (Bio101 Inc.).
  • oligonucleotides were synthesized. For this purpose, first known DNA sequences encoding for fiber protein of alternative serotypes were aligned to identify conserved regions in both the tail region as well as the knob region of the fiber protein. From the alignment, which contained the nucleotide sequence of 19 different serotypes representing all 6 subgroups, (degenerate) oligonucleotides were synthesized (see Table I, SEQ ID NOS:1-13). Also shown in Table 3 is the combination of oligonucleotides used to amplify the DNA encoding fiber protein of a specific serotype.
  • the amplification reaction (50 ⁇ l) contained 2 mM dNTPs, 25 pmol of each oligonucleotide, standard 1 ⁇ PCR buffer, 1.5 mM MgCl 2 , and 1 Unit Pwo heat stable polymerase (Boehringer Mannheim) per reaction.
  • the cycler program contained 20 cycles, each consisting of 30 seconds at 94° C., 60 seconds at 60-64° C., and 120 seconds at 72° C.
  • One-tenth of the PCR product was run on an agarose gel to demonstrate that a DNA fragment was amplified. From each different template, two independent PCR reactions were performed.
  • an AvrII fragment from the pBr/AdBamRFib16 (ECACC deposit number 01121710), pBr/AdBamRFib28, pBr/AdBamRFib40-L constructs was subcloned into the vector pBr/Ad.Bam-rITR.pac#8 (ECACC deposit #P97082121) replacing the corresponding sequences in this vector.
  • pBr/Ad.Bam-rITR.pac#8 has the same adenoviral insert as pBr/Ad.Bam-rITR but has a PacI site near the rITR that enables the ITR to be separated from the vector sequences.
  • the constuct pWE/Ad.AflII-Eco was generated as follows. PWE.pac was digested with ClaI and the 5 prime protruding ends were filled in with Klenow enzyme. The DNA was then digested with PacI and isolated from agarose gel. PWE/AflIIrITR was digested with EcoRI and after treatment with Klenow enzyme digested with PacI. The large 24 kb fragment containing the adenoviral sequences was isolated from agarose gel and ligated to the ClaI digested and blunted pWE.Pac vector. Use was made of the ligation express kit from Clontech.
  • PWE/Ad.AlfII-Eco contains Ad5 sequences from basepairs 3534-27336.
  • Three constructs, pCArchitectl-Luc ( FIG. 5 ) digested with SalI, pWE/Ad.AflII-Eco digested with PacI and EcoRI and pBr/AdBamR.pac/fibXX digested with BamHI and PacI were transfected into adenovirus producer cells (PER.C6, Fallaux et al., 1998).
  • FIG. 6 schematically depicts the method and fragments used to generate the chimeric viruses.
  • pBr/Ad.BamRfib12 was used without subcloning in the PacI containing vector and therefor was not liberated from vector sequences using PacI but was digested with ClaI which leaves approximately 160 bp of vector sequences attached to the right ITR. Furthermore, the pBr/Ad.BamRfib12 and pBr/Ad.BamRfib28 contain an internal BamHI site in the fiber sequences and were therefor digested with SalI which cuts in the vector sequences flanking the BamHI site.
  • pCLIPsal-Luc For transfection, 2 ⁇ g of pCLIPsal-Luc, and 4 ⁇ g of both pWE/Ad.AflII-Eco and pBr/AdBamR.pac/fibXX were diluted in serum free DMEM to 100 ⁇ l total volume.
  • 100 ⁇ l 2.5 ⁇ diluted lipofectamine (Gibco) in serum-free medium were added. After 30 minutes at room temperature, the DNA-lipofectamine complex solution was added to 2.5 ml of serum-free DMEM which was subsequently added to a T25 cm 2 tissue culture flask.
  • This flask contained PER.C6 cells that were seeded 24-hours prior to transfection at a density of 1 ⁇ 10 6 cells/flask. Two hours later, the DNA-lipofectamine complex containing medium was diluted once by the addition of 2.5 ml DMEM supplemented with 20% fetal calf serum. Again 24 hours later, the medium was replaced by fresh DMEM supplemented with 10% fetal calf serum. Cells were cultured for 6-8 days, subsequently harvested, and freeze/thawed 3 times. Cellular debris was removed by centrifugation for 5 minutes at 3000 rpm at room temperature.
  • PER.C6 cells T80 cm 2 tissue culture flasks. This re-infection results in full cytopathogenic effect (CPE) after 5-6 days after which the adenovirus is harvested as described above.
  • the solution was subsequently incubated for 15 minutes at 37° C. to completely crack the cells. After homogenizing the solution, 1875 ⁇ l (1M) MgCl 2 was added and 5 ml 100% glycerol. After the addition of 375 ⁇ l DNase (10 mg/ml) the solution was incubated for 30 minutes at 37° C. Cell debris was removed by centrifugation at 1880 ⁇ g for 30 minutes at room temperature without the brake on. The supernatant was subsequently purified from proteins by loading on 10 ml of freon. Upon centrifugation for 15 minutes at 2000 rpm without brake at room temperature, three bands are visible of which the upper band represents the adenovirus.
  • This band was isolated by pipetting after which it was loaded on a Tris/HCl (1M) buffered cesium chloride blockgradient (range: 1.2 to 1.4 g/ml). Upon centrifugation at 21000 rpm for 2.5 hours at 10° C., the virus was purified from remaining protein and cell debris since the virus in contrast to the other components, does not migrate into the 1.4 g/ml cesium chloride solution. The virus band is isolated after which a second purification using a Tris/HCl (1M) buffered continues gradient of 1.33 g/ml of cesium chloride is performed. After virus loading on top of this gradient, the virus is centrifuged for 17 hours at 55000 rpm at 10° C.
  • Tris/HCl (1M) buffered cesium chloride blockgradient range: 1.2 to 1.4 g/ml
  • the virus band is isolated and after the addition of 30 ⁇ l of sucrose (50 w/v) excess cesium chloride is removed by three rounds of dialysis, each round comprising 1 hour.
  • the virus is transferred to dialysis slides (Slide-a-lizer, cut off 10000 kDa, Pierce, USA).
  • the buffers used for dialysis are PBS which are supplemented with an increasing concentration of sucrose (round 1 to 3: 30 ml, 60 ml, and 150 ml sucrose (50% w/v)/1.5 liter PBS, all supplemented with 7.5 ml 2% (w/v) CaMgCl 2 ).
  • the virus is removed from the slide-a-lizer after which it is aliquoted in portions of 25 and 100 ⁇ l upon which the virus is stored at ⁇ 85° C.
  • 50 ⁇ l of the virus batch is run on a high pressure liquid chromatograph (HPLC) as described by Shamram et al. (1997).
  • Ad5.Luc The virus titers were found to be in the same range as the Ad5.Luc virus batch (Ad5.Luc: 2.2 ⁇ 10 11 vp/ml; Ad5.LucFib12: 1.3 ⁇ 10 11 vp/ml; Ad5.LucFib16: 3.1 ⁇ 10 12 vp/ml; Ad5.LucFib28: 5.4 ⁇ 10 10 vp/ml; Ad5.LucFib40-L: 1.6 ⁇ 10 12 vp/ml).
  • the protein lysate was subsequently tested for the presence of transgene expression (luciferase activity) and the protein concentration was determined to express the luciferase activity per ⁇ g of protein.
  • the results Shown in Table II, demonstrate that in contrast to the Adenovirus serotype 5 control, none of the fiber chimeras are targeted specifically to the liver or to the spleen.
  • This experiment shows that it is possible to circumvent the uptake of adenoviruses by the liver by making use of fibers of other serotypes. It also shows that the uptake by the liver is not correlated with the length of the fiber shaft, or determined solely by the ability of fiber knob to bind to CAR.
  • the fibers used have different shaft lengths and, except for fiber 16, are derived from subgroups known to have a fiber that can bind CAR (Roelvink et al. 1998).
  • Human endothelial cells were isolated, cultured and characterized as described previously (Jaffe et al. 1973; Wijnberg et al. 1997). Briefly, cells were cultured on gelatin-coated dishes in M199 supplemented with 20 mM HEPES, pH 7.3 (Flow Lab., Irvine, Scotland), 10% (v/v) human serum (local blood bank), 10% (v/v) heat-inactivated newborn calf serum (NBCS) (GIBCO BRL, Gaithersburg, Md.), 150 ⁇ g/ml crude endothelial cell growth factor, 5 U/ml heparin (Leo Pharmaceutics Products, Weesp, The Netherlands), penicillin (100 IU/ml)/streptomycin (100 ⁇ g/ml) Boehringer Mannheim, Mannheim, Del.) at 37° C.
  • HUVEC cells a pool from 4 different individuals
  • DMEM fetal bovine serum
  • FGS fetal bovine serum
  • the viruses used were besides the control Ad5, the fiber chimeras 12, 16, 28, and 40-L (each infection in triplicate). Two hours after addition of the virus the medium was replaced by normal medium.
  • the result of the transgene expression (RLU) per microgram protein determined 48 hours after infection shows that the fiber 16 chimeric adenovirus is also better suited to infect HUVEC cells even when these cells are 100% confluent which better mimics an in vivo situation. Since the Luciferase marker gene does not provide information concerning the number of cells infected another experiment was performed with adenovirus serotype 5 and the fiber 16 chimera, both carrying a green fluorescent protein (GFP) as a marker gene. This protein expression can be detected using a flow cytometer which renders information about the percentage of cells transduced as well as fluorescence per cell.
  • GFP green fluorescent protein
  • adenovirus serotype 5 for the infection of human smooth muscle cells.
  • FIG. 8 a results are shown of the transgene expression per microgram total protein after infection of HUVsmc cells. These results show that fiber chimeras 12 and 28 are unable to infect HUVsmc cells, that 40-L infects HUVsmc with similar efficiency as the control Ad5 virus, and that fiber chimera 16 infects HUVsmc significantly better.
  • the shaft and knob of fiber 16 are derived from adenovirus serotype 16 which, as described earlier, belongs to subgroup B. Based on hemagglutination assays, DNA restriction patterns, and neutralization assays, the subgroup B viruses have been further subdivided into subgroup B1 and B2 (Wadell et al. 1984).
  • Subgroup B1 members include serotypes 3, 7, 16, 21, and 51.
  • Subgroup B2 members include 11, 14, 34, and 35.
  • HUVsmc were infected with increasing amounts of virus particles per cell (156, 312, 625, 1250, 2500, 5000).
  • the fiber mutants all carry the Luciferase marker gene (Ad5Fib11.Luc: 1.1 ⁇ 10 12 vp/ml; Ad5Fib35Luc: 1.4 ⁇ 10 12 vp/ml; Ad5Fib35Luc: 1.4 ⁇ 10 12 vp/ml; Ad5Fib51Luc: 1.0 ⁇ 10 12 vp/ml).
  • efficient infection of SMC is not a general trade of all subgroup B fiber molecules.
  • fiber 16 and fiber 11 are better suited for infection of SMC than fiber 35 and fiber 51. Nevertheless, all subgroup B fiber mutants tested infect SMC better than Ad5.
  • the viruses were Ad5.LacZ (2.2 ⁇ 10 12 vp/ml), the fiber 16 chimera: Ad5Fib16.LacZ (5.2 ⁇ 10 11 vp/ml), and a fiber 51 chimera: Ad5Fib51.LacZ (2.1 ⁇ 10 12 vp/ml).
  • the pieces of saphenous vein were macroscopically photographed using a digital camera. Based on LacZ transgene expression obtained after 2 hours of virus exposure on saphenous vein slices, both the fiber 16 and the fiber 51 chimeric viruses give higher infection since much more blue staining is observed using these viruses as compared to Ad5.LacZ ( FIG. 8 e ).
  • Ad5.LucFib16 infects umbilical vein smooth muscle cells approximately 8 fold better than umbilical vein endothelial cells whereas in case of Ad5.Luc viruses endothelial cells are better infected than smooth muscle cells.
  • Ad5 infection correlated with receptor expression of these cells the presence of CAR and ⁇ -integrins was assayed on a flow cytometer. For this purpose 1 ⁇ 10 5 HUVEC cells or HUVsmc were washed once with PBS/0.5% BSA after which the cells were pelleted by centrifugation for 5 minutes at 1750 rpm at room temperature.
  • A549 cells were infected to establish whether an equal amount of virus particles of the different chimeric adenoviruses show significant differences in transgene expression on cell lines that are easily infected by adenovirus. This is to investigate whether the observed differences in infection efficiency on endothelial and smooth muscle cells are cell type specific.

Abstract

A gene delivery vehicle having been provided with at least a tissue tropism for cells selected from the group of smooth muscle cells, endothelial cells, and/or liver cells. The tissue tropism is generally provided by a virus capsid, such as one comprising protein fragments from at least two different viruses, such as two different adenoviruses, including adenovirus of subgroup C or subgroup B (for example, adenovirus 16). The protein fragments can comprise a tissue tropism-determining fragment of a fiber protein derived from a subgroup B adenovirus. Also, cells for producing such gene delivery vehicles and pharmaceutical compositions containing these gene delivery vehicles are provided. Further, a method is disclosed for delivering nucleic acid to cells such as smooth muscle cells and/or endothelial cells which involves administering to the cells an adenovirus capsid having proteins from at least two different adenoviruses and wherein at least a tissue tropism-determining fragment of a fiber protein is derived from a subgroup B adenovirus. Particular constructs are also disclosed.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application is a continuation of co-pending application Ser. No. 09/444,284, filed Nov. 19, 1999, now U.S. Pat. No. ______, which is a continuation-in-part of application Ser. No. 09/348,354, filed Jul. 7, 1999, abandoned, the contents of both of which are incorporated by this reference.
  • TECHNICAL FIELD
  • The invention relates generally to biotechnology and, more particularly, to the field of molecular genetics and medicine. In particular, the present invention relates to the field of gene therapy and, more particularly, to gene therapy using adenoviruses.
  • BACKGROUND
  • In gene therapy, genetic information is usually delivered to a host cell in order to either correct (supplement) a genetic deficiency in the host cell, or to inhibit an undesired function in the host cell, or to eliminate the host cell. Of course, the genetic information can also be intended to provide the host cell with a desired function, e.g., to supply a secreted protein to treat other cells of the host, etc.
  • Many different methods have been developed to introduce new genetic information into cells. Although many different systems may work on cell lines cultured in vitro, only the group of viral vector mediated gene delivery methods seems to be able to meet the required efficiency of gene transfer in vivo. Thus, for gene therapy purposes, most of the attention is directed toward the development of suitable viral vectors. Today, most of the attention for the development of suitable viral vectors is directed toward those vectors based on adenoviruses. These adenovirus vectors can deliver foreign genetic information very efficiently to target cells in vivo. Moreover, obtaining large amounts of adenovirus vectors is for most types of adenovirus vectors not a problem. Adenovirus vectors are relatively easy to concentrate and purify. Moreover, studies in clinical trials have provided valuable information on the use of these vectors in patients.
  • A lot of reasons exist for using adenovirus vectors for the delivery of nucleic acid to target cells in gene therapy protocols. However, some characteristics of the current vectors limit their use in specific applications. For instance, endothelial cells and smooth muscle cells are not easily transduced by the current generation of adenovirus vectors. For many gene therapy applications, such as applications in the cardiovascular area, preferably these types of cells should be genetically modified. On the other hand, in some applications, even the very good in vivo delivery capacity of adenovirus vectors is not sufficient and higher transfer efficiencies are required. This is the case, for instance, when most cells of a target tissue need to be transduced.
  • The present invention was made in the course of the manipulation of adenovirus vectors. In the following section, therefore, a brief introduction to adenoviruses is given.
  • Adenoviruses:
  • Adenoviruses contain a linear double-stranded DNA molecule of approximately 36000 base pairs. The DNA molecule contains identical Inverted Terminal Repeats (ITR) of approximately 90-140 base pairs with the exact length depending on the serotype. The viral origins of replication are within the ITRs exactly at the genome ends. The transcription units are divided into early and late regions. Shortly after infection, the E1A and E1B proteins are expressed and function in transactivation of cellular and adenoviral genes. The early regions E2A and E2B encode proteins (DNA binding protein, pre-terminal protein, and polymerase) required for the replication of the adenoviral genome (reviewed in van der Vliet, 1995). The early region E4 encodes several proteins with pleiotropic functions, e.g., transactivation of the E2 early promoter, facilitating transport and accumulation of viral mRNAs in the late phase of infection and increasing nuclear stability of major late pre-mRNAs (reviewed in Leppard, 1997). The early region 3 encodes proteins that are involved in modulation of the immune response of the host (Wold et al., 1995). The late region is transcribed from one single promoter (major late promoter) and is activated at the onset of DNA replication. Complex splicing and poly-adenylation mechanisms give rise to more than 12 RNA species coding for core proteins, capsid proteins (Penton, hexon, fiber and associated proteins), viral protease and proteins necessary for the assembly of the capsid and shut-down of host protein translation (Imperiale, M. J., Akusjnarvi, G. and Leppard, K. N. (1995). Post-transcriptional control of adenovirus gene expression. In: The molecular repertoire of adenoviruses I. P139-171. W. Doerfler and P. Bohm (eds), springer-Verlag Berlin Heidelberg).
  • Interaction between Virus and Host Cell:
  • The interaction of the virus with the host cell has mainly been investigated with the serotype C viruses Ad2 and Ad5. Binding occurs via interaction of the knob region of the protruding fiber with a cellular receptor. The receptor for Ad2 and Ad5 and probably more adenoviruses is known as the “Coxsackievirus and Adenovirus Receptor” or CAR protein (Bergelson et al., 1997). Internalization is mediated through interaction of the RGD sequence present in the penton base with cellular integrins (Wickham et al., 1993). This may not be true for all serotypes, for example, serotypes 40 and 41 do not contain a RGD sequence in their penton base sequence (Kidd et al., 1993).
  • The Fiber Protein:
  • The initial step for successful infection is binding of adenovirus to its target cell, a process mediated through fiber protein. The fiber protein has a trimeric structure (Stouten et al., 1992) with different lengths depending on the virus serotype (Signas et al., 1985; Kidd et al., 1993). Different serotypes have polypeptides with structurally similar N and C termini, but different middle stem regions. The first 30 amino acids at the N terminus are involved in anchoring of the fiber to the penton base (Chroboczek et al., 1995), especially the conserved FNPVYP (SEQ ID NO:______) region in the tail (Arnberg et al., 1997). The C-terminus, or knob, is responsible for initial interaction with the cellular adenovirus receptor. After this initial binding, secondary binding between the capsid penton base and cell-surface integrins leads to internalization of viral particles in coated pits and endocytosis (Morgan et al., 1969; Svensson and Persson, 1984; Varga et al., 1991; Greber et al., 1993; Wickham et al., 1993). Integrins are αβ-heterodimers of which at least 14 α-subunits and 8 β-subunits have been identified (Hynes, 1992). The array of integrins expressed in cells is complex and will vary between cell types and cellular environment. Although the knob contains some conserved regions, between serotypes, knob proteins show a high degree of variability, indicating that different adenovirus receptors exist.
  • Adenoviral Serotypes:
  • At present, six different subgroups of human adenoviruses have been proposed, which in total encompass approximately 50 distinct adenovirus serotypes. Besides these human adenoviruses, many animal adenoviruses have been identified (see, e.g., Ishibashi and Yasue, 1984). A serotype is defined on the basis of its immunological distinctiveness as determined by quantitative neutralization with animal antiserum (horse, rabbit). If neutralization shows a certain degree of cross-reaction between two viruses, distinctiveness of serotype is assumed if A) the hemagglutinins are unrelated, as shown by lack of cross-reaction on hemagglutination-inhibition, or B) substantial biophysical/biochemical differences in DNA exist (Francki et al., 1991). The serotypes identified last (42-49) were isolated for the first time from HIV infected patients (Hierholzer et al., 1988; Schnurr et al., 1993). For reasons not well understood, most of such immuno-compromised patients shed adenoviruses that were never isolated from immuno-competent individuals (Hierholzer et al., 1988, 1992; Khoo et al., 1995).
  • Besides differences towards the sensitivity against neutralizing antibodies of different adenovirus serotypes, adenoviruses in subgroup C such as Ad2 and Ad5 bind to different receptors as compared to adenoviruses from subgroup B such as Ad3 and Ad7 (Defer et al., 1990; Gall et al., 1996). Likewise, it was demonstrated that receptor specificity could be altered by exchanging the Ad3 knob protein with the Ad 5 knob protein, and vice versa (Krasnykh et al., 1996; Stevenson et al., 1995, 1997). Serotypes 2, 4, 5 and 7 all have a natural affiliation towards lung epithelia and other respiratory tissues. In contrast, serotypes 40 and 41 have a natural affiliation towards the gastrointestinal tract. These serotypes differ in at least capsid proteins (penton-base, hexon), proteins responsible for cell binding (fiber protein), and proteins involved in adenovirus replication. It is unknown to what extent the capsid proteins determine the differences in tropism found between the serotypes. It may very well be that post-infection mechanisms determine cell type specificity of adenoviruses. It has been shown that adenoviruses from serotypes A (Ad12 and Ad31), C (Ad2 and Ad5), D (Ad9 and Ad15), E (Ad4) and F (Ad41) all are able to bind labeled, soluble CAR (sCAR) protein when immobilized on nitrocellulose. Furthermore, binding of adenoviruses from these serotypes to Ramos cells, that express high levels of CAR but lack integrins (Roelvink et al., 1996), could be efficiently blocked by addition of sCAR to viruses prior to infection (Roelvink et al., 1998). However, the fact that (at least some) members of these subgroups are able to bind to CAR does not exclude that these viruses have different infection efficiencies in various cell types. For example, subgroup D serotypes have relatively short fiber shafts compared to subgroup A and C viruses. It has been postulated that the tropism of subgroup D viruses is to a large extent determined by the penton base binding to integrins (Roelvink et al., 1996; Roelvink et al., 1998). Another example is provided by Zabner et al., 1998 who tested 14 different serotypes on infection of human ciliated airway epithelia (CAE) and found that serotype 17 (subgroup D) was bound and internalized more efficiently than all other viruses, including other members of subgroup D. Similar experiments using serotypes from subgroup A-F in primary fetal rat cells showed that adenoviruses from subgroup A and B were inefficient Whereas viruses from subgroup D were most efficient (Law et. al, 1998). Also, in this case, viruses within one subgroup displayed different efficiencies. The importance of fiber binding for the improved infection of Ad17 in CAE was shown by Armentano et al. (PCT International Patent Publication WO 98/22609) who made a recombinant LacZ Ad2 virus with a fiber gene from Ad17 and showed that the chimeric virus infected CAE more efficiently than LacZ Ad2 viruses with Ad2 fibers.
  • Thus, despite their shared ability to bind CAR, differences in the length of the fiber, knob sequence and other capsid proteins, e.g., penton base of the different serotypes may determine the efficiency by which an adenovirus infects a certain target cell. Of interest in this respect is the ability of Ad5 and Ad2 fibers but not of Ad3 fibers to bind to fibronectin III and MHC class 1 α2 derived peptides. This suggests that adenoviruses are able to use cellular receptors other than CAR (Hong et al., 1997). Serotypes 40 and 41 (subgroup F) are known to carry two fiber proteins differing in the length of the shaft. The long shafted 41L fiber is shown to bind to CAR whereas the short shafted 41S is not capable of binding CAR (Roelvink et al., 1998). The receptor for the short fiber is not known.
  • Adenoviral Gene Delivery Vectors:
  • Most adenoviral gene delivery vectors currently used in gene therapy are derived from the serotype C adenoviruses Ad2 or Ad5. The vectors have a deletion in the E1 region, where novel genetic information can be introduced. The E1 deletion renders the recombinant virus replication defective. It has been demonstrated extensively that recombinant adenovirus, in particular serotype 5, is suitable for efficient transfer of genes in vivo to the liver, the airway epithelium and solid tumors in animal models and human xenografts in immuno-deficient mice (Bout 1996, 1997; Blaese et al., 1995).
  • Gene transfer vectors derived from adenoviruses (adenoviral vectors) have a number of features that make them particularly useful for gene transfer:
      • 1) the biology of the adenoviruses is well characterized;
      • 2) adenovirus is not generally associated with severe human pathology;
      • 3) the virus is extremely efficient in introducing its DNA into the host cell;
      • 4) the virus can infect a wide variety of cells and has a broad host-range;
      • 5) the virus can be produced at high titers in large quantities; and
      • 6) the virus can be rendered replication defective by deletion of the early-region 1 (E1) of the viral genome (Brody and Crystal, 1994).
  • However, there is still a number of drawbacks associated with the use of adenoviral vectors:
      • 1) Adenoviruses, especially the well investigated serotypes Ad2 and Ad5, usually elicit an immune response in the host into which they are introduced;
      • 2) it is currently not feasible to target the virus to certain cells and tissues;
      • 3) the replication and other functions of the adenovirus are not always very well suited for the cells; which are to be provided with the additional genetic material; and
      • 4) the serotypes Ad2 or Ad5 are not ideally suited for delivering additional genetic material to organs other than the liver. The liver can be particularly well transduced with vectors derived from Ad2 or Ad5.
        Delivery of such vectors via the bloodstream leads to a significant delivery of the vectors to the cells of the liver. In therapies where other cell types other than liver cells need to be transduced some means of liver exclusion must be applied to prevent uptake of the vector by these cells. Current methods rely on the physical separation of the vector from the liver cells, most of these methods rely on localizing the vector and/or the target organ via surgery, balloon angioplasty or direct injection into an organ via, for instance, needles. Liver exclusion is also being practiced through delivery of the vector to compartments in the body that are essentially isolated from the bloodstream, thereby preventing transport of the vector to the liver. Although these methods mostly succeed in avoiding gross delivery of the vector to the liver, most of the methods are crude and still have considerable leakage and/or have poor target tissue penetration characteristics. In some cases, inadvertent delivery of the vector to liver cells can be toxic to the patient. For instance, delivery of a herpes simplex virus (HSV) thymidine kinase (TK) gene for the subsequent killing of dividing cancer cells through administration of gancyclovir is quite dangerous when also a significant amount of liver cells are transduced by the vector. Significant delivery and subsequent expression of the HSV-TK gene to liver cells is associated with severe toxicity. Thus, a discrete need exists for an inherently safe vector provided with the property of a reduced transduction efficiency of liver cells.
    BRIEF SUMMARY OF THE INVENTION
  • The present invention provides gene therapy methods, compounds and medicines. The present invention is particularly useful in gene therapy applications where endothelial cells and/or smooth muscle cells form the target cell type. The present invention relates to gene delivery vehicles provided with a tissue tropism for at least endothelial cells and/or smooth muscle cells. The present invention further relates to gene delivery vehicles having been deprived of a tissue tropism for liver cells.
  • BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
  • Table I: Oligonucleotides and degenerate oligonucleotides used for the amplification of DNA encoding fiber proteins derived from alternative adenovirus serotypes. (Bold letters represent NdeI restriction site (A-E), NsiI restriction site (1-6, 8), or PacI restriction site (7).
  • Table II: Biodistribution of chimeric adenovirus upon intravenous tail vein injection. Values represent luciferase activity/μg of total protein. All values below 200 Relative light units/μg protein are considered background. ND=not determined.
  • Table III: Expression of CAR and integrins on the cell surface of endothelial cells and smooth muscle cells. 70%: Cells harvested for FACS analysis at a cell density of 70% confluency. 100%: Cells harvested for FACS analysis at a cell density of 100% confluency. PER.C6 cells were taken as a control for antibody staining. Values represent percentages of cells that express CAR or either one of the integrins at levels above background. As background control, HUVECs or HUVsmc were incubated only with the secondary, rat-anti-mouse IgG1 labeled antibody.
  • Table IV: Determination of transgene expression (luciferase activity) per μg of total cellular protein, after infection of A549 cells.
  • FIG. 1: Schematic drawing of the pBr/Ad.Bam-rITR construct.
  • FIG. 2: Schematic drawing of the strategy used to delete the fiber gene from the pBr/Ad.Bam-rITR construct.
  • FIG. 3: Schematic drawing of construct pBr/Ad.BamRΔfib (ECACC deposit number 01121708).
  • FIG. 4: Sequences of the chimeric fibers Ad5 (SEQ ID NO:16) Ad5/12 (SEQ ID NO:17), Ad5/16 (SEQ ID NO:18), Ad5/28 (SEQ ID NO:19), and Ad5/40-L (SEQ ID NO:20).
  • FIG. 5: Schematic drawing of the construct pClipsal.-Luc.
  • FIG. 6: Schematic drawing of the method to generate chimeric adenoviruses using three overlapping fragments. Early (E) and late regions (L) are indicated. L5 is the fiber coding sequence.
  • FIG. 7: A) Infection of HUVEC cells using different amounts of virus particles per cell and different fiber chimeric adenoviruses. Virus concentration: 10000 vp/cell (=white bar), 5000 vp/cell (=grey bar), 2500 vp/cell (=Black bar) 1000 vp/cell (light grey bar, 250 and 50 vp/cell no detectable luciferase activity above background. Luciferase activity is expressed in relative light units (RLU) per microgram of cellular protein. B) Infection of HUVEC cells using different concentrations of cells (22,500, 45,000, 90,000, or 135,000 cells seeded per well) and either adenovirus serotype 5 (black bar) or the fiber 16 chimeric adenovirus (white bar). Luciferase activity is expressed in RLU per microgram cellular protein. C) Flow cytometric analysis on Human aorta EC transduced with 500 (Black bar) or 5000 (grey bar) virus particles per cell of Ad5 or the fiber 16 chimeric virus (Fib16). Non-infected cells were used to set the background at 1% and a median fluorescence of 5.4. The maximum shift in the median fluorescence that can be observed on a flow cytometer is 9999. This latter indicates that at 5000 Vp/cell both Ad5 and Fib 16 are outside the sensitivity scale of the flow cytometer.
  • FIG. 8: A) Infection of HUVsmc cells using different amounts of virus particles per cell and different fiber mutant Ad5 based adenoviruses. Virus concentration: 5000 Vp/cell (=white bar), 2500 vp/cell (=grey bar), 1250 vp/cell (=dark grey bar), 250 vp/cell (=black bar), or 50 vp/cell (light grey bar). Luciferase activity is expressed as RLU per microgram cellular protein. B) Infection of HUVsmc cells using different concentrations of cells (10,000, 20,000, 40,000, 60,000, or 80,000 cells per well) and either adenovirus serotype 5 (white bars) or the fiber 16 chimeric adenovirus (black bars). A plateau is observed after infection with chimeric fiber 16 adenovirus due to the fact that transgene expression is higher than the sensitivity range of the bioluminometer used. C) Human umbilical vein SMC transduced with 500 vp/cell (black bar) or 5000 vp/cell (grey bar) using either Ad5 or the fiber 16 mutant (Fib 16). Non-transduced cells were used to set a background median fluorescence of approximately 1. Shown is the median fluorescence of GFP expression as measured by flow cytometry. D) HUVsmc were infected with 312 (light grey bar), 625 (grey bar), 1250 (black bar), 2500 (dark grey bar), 5000 (light grey bar), or 10,000 (white bar) virus particles per cell of either the fiber 11, 16, 35, or 51 chimeric virus. Luciferase transgene expression expressed as RLU per microgram protein was measured 48 hours after virus exposure. E) Macroscopic photographs of LacZ staining on saphenous samples. Nuclear targeted LacZ (ntLacZ) yields a deep blue color which appears black or dark grey in non-color prints. F) Macroscopic photographs of LacZ staining on pericard samples. Nuclear targeted LacZ (ntLacZ) gives a deep blue color which appears black in non-color prints. G) Macroscopic photographs of LacZ staining on right coronary artery samples. Nuclear targeted LacZ (ntLacZ) gives a deep blue color which appears black in non-color prints. H) LacZ staining on Left artery descending (LAD) samples. Nuclear targeted LacZ (ntLacZ) gives a deep blue color which appears black in non-color prints.
  • FIG. 9: Sequences including the gene encoding adenovirus 16 fiber protein as published in Genbank (SEQ ID NO:21) and sequence including a gene encoding a fiber from an adenovirus 16 variant as isolated in the present invention, wherein the sequences of the fiber protein are from the NdeI-site (SEQ ID NO:22). FIG. 9A is a nucleotide sequence comparison (SEQ ID NOS:21 (upper stand) and 22 (lower strand)). FIG. 9B is an amino-acid comparison (SEQ ID NOS:23 (upper strand) and 24 (lower strand)).
  • DETAILED DESCRIPTION OF THE INVENTION
  • The current invention provides materials and methods to overcome the limitations of adenoviral vectors mentioned above. In a broad sense, the invention provides new adenoviruses, derived in whole or in part from serotypes different from Ad5. Specific genes of serotypes with preferred characteristics may be combined in a chimeric vector to give rise to a vector that is better suited for specific applications. Preferred characteristics include, but are not limited to, improved infection of a specific target cell, reduced infection of non-target cells, improved stability of the virus, reduced uptake in antigen presenting cells (APC), or increased uptake in APC, reduced toxicity to target cells, reduced neutralization in humans or animals, reduced or increased CTL response in humans or animals, better and/or prolonged transgene expression, increased penetration capacity in tissues, improved yields in packaging cell lines, etc.
  • One aspect of the present invention facilitates the combination of the low immunogenicity of some adenoviruses with the characteristics of other adenoviruses that allow efficient gene therapy. Such characteristics may be a high specificity for certain host cells, a good replication machinery for certain cells, a high rate of infection in certain host cells, low infection efficiency in non-target cells, high or low efficiency of APC infection, etc. The invention thus may provide chimeric adenoviruses having the useful properties of at least two adenoviruses of different serotypes.
  • Typically, two or more requirements from the above non-exhaustive list are required to obtain an adenovirus capable of efficiently transferring genetic material to a host cell. Therefore, the present invention provides adenovirus vectors that can be used as cassettes to insert different adenoviral genes from different adenoviral serotypes at the required sites. This way, one can obtain a vector capable of producing a chimeric adenovirus, whereby of course also a gene of interest can be inserted (for instance at the site of E1 of the original adenovirus). In this manner, the chimeric adenovirus to be produced can be adapted to the requirements and needs of certain hosts in need of gene therapy for certain disorders. To enable this virus production, a packaging cell will generally be needed in order to produce sufficient amount of safe chimeric adenoviruses.
  • In one of its aspects, the present invention provides adenoviral vectors comprising at least a fragment of a fiber protein of an adenovirus from subgroup B. This fiber protein may be the native fiber protein of the adenoviral vector or may be derived from a serotype different from the serotype the adenoviral vector is based on. In the latter case, the adenoviral vector according to the invention is a chimeric adenovirus displaying at least a fragment of the fiber protein derived from subgroup B adenovirus, wherein the fragment comprises at least the receptor binding sequence. Typically such a virus will be produced using a vector (typically a plasmid, a cosmid or baculovirus vector). Such vectors are also subject of the present invention. A preferred vector is a vector that can be used to make a chimeric recombinant virus specifically adapted to the host to be treated and the disorder to be treated.
  • The present invention also provides a chimeric adenovirus based on adenovirus type 5, but having at least a fragment of the fiber sequence from adenovirus type 16, wherein the fragment of the fiber of Ad16 comprises the fragment of the fiber protein that is involved in binding a host cell. The present invention also provides chimeric adenoviral vectors that show improved infection as compared to adenoviruses from other subgroups in specific host cells, for example, but not limited to, endothelial cells and smooth muscle cells of human or animal origin. An important feature of the present invention is the means to produce the chimeric virus. Typically, one does not want an adenovirus batch to be administered to the host cell, which contains replication competent adenovirus. In general, therefore, it is desired to omit one or more genes from the adenoviral genome on the vector encoding the chimeric virus and to supply these genes in the genome of the cell in which the vector is brought to produce chimeric adenovirus. Such a cell is usually called a “packaging cell.” The invention thus also provides a packaging cell for producing a chimeric adenovirus according to the invention, comprising, in trans, all elements necessary for adenovirus production not present on the adenoviral vector according to the invention. Typically, vector and packaging cell are adapted to one another in that they have all the necessary elements, but that they do not have overlapping elements which lead to replication competent virus by recombination. Thus, the invention also provides a kit of parts comprising a packaging cell according to the invention and a recombinant vector according to the invention wherein there is essentially no sequence overlap leading to recombination resulting in the production of replication competent adenovirus between the cell and the vector. For certain applications, for example when the therapy is aimed at eradication of tumor cells, adenoviral vector according to the invention may be replication competent or capable of replicating under certain conditions, for example, in specific cell types like tumor cells or tumor endothelial cells.
  • It is within the scope of the invention to insert more genes, or a functional part of these genes from the same or other serotypes into the adenoviral vector replacing the corresponding native sequences. Thus, for example, replacement of (or a functional part of the) fiber sequences with corresponding sequences of other serotypes may be combined with, for example, replacements of (or a functional part of) other capsid genes like penton base or hexon with corresponding sequences of the serotype or of other distinct serotypes. Persons skilled in the art will understand that other combinations not limited to genes are possible and within the scope of the invention.
  • The chimeric adenoviral vector according to the invention may originate from at least two different serotypes. This may provide the vector with preferred characteristics such as improved infection of target cells and/or less infection of non-target cells, improved stability of the virus, reduced immunogenicity in humans or animals (e.g., reduced uptake in APC, reduced neutralization in the host and/or reduced cytotoxic T-lymphocyte (CTL) response), increased penetration of tissue, better longevity of transgene expression, etc. In this aspect, it is preferred to use capsid genes, e.g., penton and/or hexon genes from less immunogenic serotypes as defined by the absence or the presence of low amounts of neutralizing antibodies in the vast majority of hosts.
  • It is also preferred to use fiber and/or penton sequences from serotypes that show improved binding and internalization in the target cells. Furthermore, it is preferred to delete from the viral vector those genes which lead to expression of adenoviral genes in the target cells. In this aspect, a vector deleted of all adenoviral genes is also preferred. Furthermore, it is preferred that the promoter driving the gene of interest to be expressed in the target cells is a cell type specific promoter.
  • In order to be able to precisely adapt the viral vector and provide the chimeric virus with the desired properties at will, it is preferred that a library of adenoviral genes be provided wherein the genes to be exchanged are located on plasmid- or cosmid-based adenoviral constructs wherein the genes or the sequences to be exchanged are flanked by restriction sites. The preferred genes or sequences can be selected from the library and inserted in the adenoviral constructs that are used to generate the viruses. Typically, such a method comprises a number of restriction and ligation steps and transfection of a packaging cell. The adenoviral vector can be transfected in one piece, or as two or more overlapping fragments, wherein viruses are generated by homologous recombination. For example, the adenoviral vector may be built up from two or more overlapping sequences for insertion or replacements of a gene of interest in, for example, the E1 region, for insertion or replacements in penton and/or hexon sequences, and for insertions or replacements into fiber sequences. Thus, the invention provides a method for producing chimeric adenoviruses having one or more desired properties like a desired host range and diminished antigenicity, comprising providing one or more vectors according to the invention having the desired insertion sites, inserting into these vectors at least a functional part of a fiber protein derived from an adenovirus serotype having the desired host range and/or inserting a functional part of a capsid protein derived from an adenovirus serotype having a relatively low antigenicity and transfecting these vectors in a packaging cell according to the invention and allowing for production of chimeric viral particles. Of course, other combinations of other viral genes originating from different serotypes can also be inserted as disclosed herein before. Chimeric viruses having only one non-native sequence in addition to an insertion or replacement of a gene of interest in the E1 region, are also within the scope of the invention.
  • An immunogenic response to adenovirus that typically occurs is the production of neutralizing antibodies by the host. This response is typically a reason for selecting a capsid protein like penton, hexon and/or fiber of a less immunogenic serotype.
  • Of course, it may not be necessary to make chimeric adenoviruses that have complete proteins from different serotypes. It is well within the skill of the art to produce chimeric proteins, for instance in the case of fiber proteins it is very well possible to have the base of one serotype and the shaft and the knob from another serotype. In this manner, it becomes possible to have the parts of the protein responsible for assembly of viral particles originate from one serotype, thereby enhancing the production of intact viral particles. Thus, the invention also provides a chimeric adenovirus wherein the hexon, penton, fiber and/or other capsid proteins are chimeric proteins originating from different adenovirus serotypes. Besides generating chimeric adenoviruses by swapping entire wild type capsid (protein) genes etc. or parts thereof, it is also within the scope of the present invention to insert capsid (protein) genes etc. carrying non-adenoviral sequences or mutations, such as point mutations, deletions, insertions, etc., which can be easily screened for preferred characteristics such as temperature stability, assembly, anchoring, redirected infection, altered immune response etc. Again, other chimeric combinations can also be produced and are within the scope of the present invention.
  • It has been demonstrated in mice and rats that upon in vivo systemic delivery of recombinant adenovirus of commonly used serotypes for gene therapy purposes more than 90% of the virus is trapped in the liver, (Herz et al., 1993; Kass-Eisler et al., 1994; Huard et al., 1995). It is also known that human hepatocytes are efficiently transduced by adenovirus serotype 5 vectors (Castell, J. V., Hernandez, D. Gomez-Foix, A. M., Guillen, I, Donato, T. and Gomez-Lechon, M. J. (1997). Adenovirus-mediated gene transfer into human hepatocytes: analysis of the biochemical functionality of transduced cells. Gene Ther. 4 (5), p 455-464). Thus, in vivo gene therapy by systemic delivery of Ad2 or Ad5 based vectors is seriously hampered by the efficient uptake of the viruses in the liver leading to unwanted toxicity and less virus being available for transduction of the target cells. Therefore, alteration of the adenovirus serotype 5 host cell range to be able to target other organs in vivo is a major interest of the invention.
  • To obtain redirected infection of recombinant adenovirus serotype 5, several approaches have been or still are under investigation. Wickham et al. have altered the RGD (Arg, Gly, Asp) motif in the penton base which is believed to be responsible for the αβ3 and αβ5 integrin binding to the penton base. They have replaced this RGD motif by another peptide motif which is specific for the α4β1 receptor. In this way, targeting the adenovirus to a specific target cell could be accomplished (Wickham et al., 1995). Krasnykh et al. (1998) have made use of the HI loop available in the knob. This loop is, based on X-ray crystallography, located on the outside of the knob trimeric structure and therefore is thought not to contribute to the intramolecular interactions in the knob. Insertion of a FLAG coding sequence into the HI loop resulted in fiber proteins that were able to trimerize and it was further shown that viruses containing the FLAG sequence in the knob region could be made. Although interactions of the FLAG-containing knob with CAR are not changed, insertion of ligands in the HI loop may lead to retargeting of infection. Although successful introduction of changes in the adenovirus serotype 5 fiber and penton-base have been reported, the complex structure of knob and the limited knowledge of the precise amino acids interacting with CAR render such targeting approaches laborious and difficult. The use of antibodies binding to CAR and to a specific cellular receptor has also been described (Wickham et al., 1996; Rogers et al., 1997). This approach is however limited by the availability of specific antibody and by the complexity of the gene therapy product.
  • To overcome the limitations described above, we used pre-existing adenovirus fibers, penton base proteins, hexon proteins or other capsid proteins derived from other adenovirus serotypes. By generating chimeric adenovirus serotype 5 libraries containing structural proteins of alternative adenovirus serotypes, we have developed a technology, which enables rapid screening for a recombinant adenoviral vector with preferred characteristics.
  • The present invention provides methods for the generation of chimeric capsids which can be targeted to specific cell types in vitro as well as in vivo, and thus have an altered tropism for certain cell types. The present invention further provides methods and means by which an adenovirus or an adenovirus capsid can be used as a protein or nucleic acid delivery vehicle to a specific cell type or tissue.
  • The generation of chimeric adenoviruses based on adenovirus serotype 5 with modified late genes is described. For this purpose, three plasmids, which together contain the complete adenovirus serotype 5 genome, were constructed. From one of these plasmids part of the DNA encoding the adenovirus serotype 5 fiber protein was removed and replaced by linker DNA sequences that facilitate easy cloning. This plasmid subsequently served as template for the insertion of DNA encoding fiber protein derived from different adenovirus serotypes. The DNA sequences derived from the different serotypes were obtained using the polymerase chain reaction technique in combination with (degenerate) oligonucleotides. At the former E1 location in the genome of adenovirus serotype 5, any gene of interest can be cloned. A single transfection procedure of the three plasmids together results in the formation of a recombinant chimeric adenovirus. Alternatively, cloning of the sequences obtained from the library of genes can be such that the chimeric adenoviral vector is built up from one or two fragments. For example, one construct contains at least the left ITR and sequences necessary for packaging of the virus, an expression cassette for the gene of interest and sequences overlapping with the second construct comprising all sequences necessary for replication and virus formation not present in the packaging cell as well as the non-native sequences providing the preferred characteristics. This new technology of libraries consisting of chimeric adenoviruses thus allows for a rapid screening improved recombinant adenoviral vectors for in vitro and in vivo gene therapy purposes.
  • The use of adenovirus type 5 for in vivo gene therapy is limited by the apparent inability to infect certain cell types, e.g., human endothelial cells and smooth muscle cells and the preference of infection of certain organs, e.g., liver and spleen. Specifically, this has consequences for treatment of cardiovascular diseases like restenosis and pulmonary hypertension. Adenovirus-mediated delivery of human ceNOS (constitutive endothelial nitric oxide synthase) has been proposed as treatment for restenosis after percutaneous transluminal coronary angioplasty (PTCA). Restenosis is characterized by progressive arterial remodeling, extracellular matrix formation and intimal hyperplasia at the site of angioplasty (Schwartz et al., 1993; Carter et al., 1994; Shi et al., 1996). NO is one of the vasoactive factors shown to be lost after PTCA-induced injury to the endothelial barrier (Lloyd Jones and Bloch, 1996). Thus, restoration of NO levels after balloon-induced injury by means of adenoviral delivery of ceNOS may prevent restenosis (Varenne et al., 1998). Other applications for gene therapy whereby the viruses or chimeric viruses according to the invention are superior to Ad2 or Ad5 based viruses, given as non-limiting examples, are production of proteins by endothelial cells that are secreted into the blood, treatment of hypertension, preventive treatment of stenosis during vein grafting, angiogenesis, heart failure, renal hypertension and others.
  • In one embodiment, this invention includes adenoviral vectors that are, amongst others, especially suited for gene delivery to endothelial cells and smooth muscle cells important for treatment of cardiovascular disorders. The adenoviral vectors preferably are derived from subgroup B adenoviruses or contain at least a functional part of the fiber protein from an adenovirus from subgroup B comprising at least the cell-binding moiety of the fiber protein. In a further preferred embodiment, the adenoviral vectors are chimeric vectors based on adenovirus type 5 and contain at least a functional part of the fiber protein from adenovirus type 16.
  • In another embodiment, this invention provides adenoviral vectors or chimeric adenoviral vectors that escape the liver following systemic administration. Preferably, these adenoviral vectors are derived from subgroup A, B, D, or F in particular serotypes 12, 16, 28 and 40 or contain at least the cell-binding moiety of the fiber protein derived from the adenoviruses.
  • It is to be understood that in all embodiments, the adenoviral vectors may be derived from the serotype having the desired properties or that the adenoviral vector is based on an adenovirus from one serotype and contains the sequences comprising the desired functions of another serotype, these sequences replacing the native sequences in the serotype.
  • In another aspect, this invention describes chimeric adenoviruses and methods to generate these viruses that have an altered tropism different from that of adenovirus serotype 5. For example, viruses based on adenovirus serotype 5 but displaying any adenovirus fiber existing in nature. This chimeric adenovirus serotype 5 is able to infect certain cell types more efficiently, or less efficiently in vitro and in vivo than the adenovirus serotype 5. Such cells include, but are not limited to, endothelial cells, smooth muscle cells, dendritic cells, neuronal cells, glial cells, synovical cells, lung epithelia cells, hemopoietic stem cells, monocytic/macrophage cells, tumor cells, skeletal muscle cells, mesothelial cells, synoviocytes, etc.
  • In another aspect, the invention describes the construction and use of libraries consisting of distinct parts of adenovirus serotype 5 in which one or more genes or sequences have been replaced with DNA derived from alternative human or animal serotypes. This set of constructs, in total encompassing the complete adenovirus genome, allows for the construction of unique chimeric adenoviruses customized for a certain disease, group of patients or even a single individual.
  • In all aspects of the invention, the chimeric adenoviruses may, or may not, contain deletions in the E1 region and insertions of heterologous genes linked either or not to a promoter. Furthermore, chimeric adenoviruses may, or may not, contain deletions in the E3 region and insertions of heterologous genes linked to a promoter. Furthermore, chimeric adenoviruses may, or may not, contain deletions in the E2 and/or E4 region and insertions of heterologous genes linked to a promoter. In the latter case, E2 and/or E4 complementing cell lines are used to generate recombinant adenoviruses. In fact, any gene in the genome of the viral vector can be taken out and supplied in trans. Thus, in the extreme, chimeric viruses do not contain any adenoviral genes in their genome and are by definition situation minimal adenoviral vectors. In this case all adenoviral functions are supplied in trans using stable cell lines and/or transient expression of these genes. A method for producing minimal adenoviral vectors is described in PCT International Publication WO97/00326 and is incorporated by reference herein. In another case Ad/AAV chimeric molecules are packaged into the adenovirus capsids of the invention. A method for producing Ad/AAV chimeric vectors is described in European Patent Office publication EP 1 042 494 and is incorporated by reference herein. In principle, any nucleic acid may be provided with the adenovirus capsids of the invention.
  • In one embodiment, the invention provides a gene delivery vehicle having been provided with at least a tissue tropism for smooth muscle cells and/or endothelial cells. In another embodiment, the invention provides a gene delivery vehicle deprived of a tissue tropism for at least liver cells. Preferably, the gene delivery vehicle is provided with a tissue tropism for at least smooth muscle cells and/or endothelial cells and deprived of a tissue tropism for at least liver cells. In a preferred embodiment, the gene delivery vehicle is provided with a tissue tropism for at least smooth muscle cells and/or endothelial cells and/or deprived of a tissue tropism for at least liver cells using a fiber protein derived from a subgroup B adenovirus, preferably of adenovirus 16. In a preferred aspect of the invention, the gene delivery vehicle comprises a virus capsid. Preferably, this virus capsid comprises a virus capsid derived in whole or in part from an adenovirus of subgroup B, preferably from adenovirus 16, or it comprises proteins, or parts thereof, from an adenovirus of subgroup B, preferably of adenovirus 16. In a preferred embodiment, the virus capsid comprises proteins, or fragments thereof, from at least two different viruses, preferably adenoviruses. In a preferred embodiment of this aspect of the invention, at least one of the viruses is an adenovirus of subgroup B, preferably adenovirus 16.
  • In a preferred embodiment, the gene delivery vehicle comprises an adenovirus fiber protein or fragments thereof. The fiber protein is preferably derived from an adenovirus of subgroup B, preferably adenovirus 16. The gene delivery vehicle may further comprise other fiber proteins, or fragments thereof, from other adenoviruses. The gene delivery vehicle may or may not comprise other adenovirus proteins. Nucleic acid may be linked directly to fiber proteins, or fragments thereof, but may also be linked indirectly. Examples of indirect linkages include, but are not limited to, packaging of nucleic acid into adenovirus capsids or packaging of nucleic acid into liposomes, wherein a fiber protein, or a fragment thereof, is incorporated into an adenovirus capsid or linked to a liposome. Direct linkage of nucleic acid to a fiber protein, or fragment thereof, may be performed when the fiber protein, or a fragment thereof, is not part of a complex or when the fiber protein or a fragment thereof, is part of a complex, such as an adenovirus capsid.
  • In one embodiment, a gene delivery vehicle is provided comprising an adenovirus fiber protein wherein the fiber protein comprises a tissue-determining fragment of an adenovirus of subgroup B adenovirus preferably of adenovirus 16. Adenovirus fiber protein comprises three functional domains. One domain, the base, is responsible for anchoring the fiber to a penton base of the adenovirus capsid. Another domain, the knob, is responsible for receptor recognition whereas the shaft domain functions as a spacer separating the base from the knob. The different domains may also have other functions. For instance, the shaft is presumably also involved in target cell specificity. Each of the domains mentioned above may be used to define a fragment of a fiber. However, fragments may also be identified in another way. For instance, the knob domain comprises a receptor binding fragment and a shaft binding fragment. The base domain comprises of a penton base binding fragment and a shaft binding fragment. Moreover, the shaft comprises repeated stretches of amino acids. Each of these repeated stretches may be a fragment.
  • Fiber proteins possess tissue tropism-determining properties. The most well described fragment of the fiber protein involved in tissue tropism is the knob domain. However, the shaft domain of the fiber protein also possesses tissue tropism-determining properties. However, not all of the tissue tropism-determining properties of an adenovirus capsid are incorporated into a fiber protein.
  • A “tissue tropism-determining fragment” of a fiber protein may be a single fragment of a fiber protein or a combination of fragments of at least one fiber protein, wherein the tissue tropism-determining fragment, either alone or in combination with a virus capsid, determines the efficiency with which a gene delivery vehicle can transduce a given cell or cell type, preferably but not necessarily in a positive way. With a “tissue tropism for liver cells” is meant a tissue tropism for cells residing in the liver, preferably liver parenchyma cells.
  • A tissue tropism for a certain tissue may be provided by increasing the efficiency with which cells of the tissue are transduced, alternatively, a tissue tropism for a certain tissue may be provided by decreasing the efficiency with which other cells than the cells of the tissue are transduced.
  • In a preferred embodiment, a fiber protein derived from a subgroup B adenovirus, preferably adenovirus 16, is combined with the non-fiber capsid proteins from an adenovirus of subgroup C, preferably of adenovirus 5.
  • In one aspect of the invention, a gene delivery vehicle is provided comprising a nucleic acid derived from an adenovirus. In a preferred embodiment of the invention the adenovirus nucleic acid comprises at least one nucleic acid sequence encoding a fiber protein comprising at least a tissue tropism-determining fragment of a subgroup B adenovirus fiber protein, preferably of adenovirus 16. In a preferred aspect, the adenovirus comprises nucleic acid from at least two different adenoviruses. In a preferred aspect, the adenovirus comprises nucleic acid from at least two different adenoviruses wherein at least one nucleic acid sequence encoding a fiber protein comprises at least a tissue tropism-determining fragment of a subgroup B adenovirus fiber protein, preferably of adenovirus 16.
  • In a preferred embodiment, the adenovirus nucleic acid is modified such that the capacity of the adenovirus nucleic acid to replicate in a target cell has been reduced or disabled. This may be achieved through, for example, inactivating or deleting genes encoding early region proteins.
  • In another preferred embodiment, the adenovirus nucleic acid is modified such that the capacity of a host immune system to mount an immune response against adenovirus proteins encoded by the adenovirus nucleic acid has been reduced or disabled. This may be achieved through deletion of genes encoding proteins of early region 2 and/or early region 4. Alternatively, genes encoding early region 3 proteins may be deleted, or on the contrary, considering the anti-immune system function of some of the proteins encoded by the genes in early region 3, the expression of early region 3 proteins may be enhanced for some purposes. Also, the adenovirus nucleic acid may be altered by a combination of two or more of the specific alterations of the adenovirus nucleic acid mentioned above. It is clear that when essential genes are deleted from the adenovirus nucleic acid, the genes must be complemented in the cell that is going to produce the adenovirus nucleic acid, the adenovirus vector, the vehicle or the chimeric capsid. The adenovirus nucleic acid may also be modified such that the capacity of a host immune system to mount an immune response against adenovirus proteins encoded by the adenovirus nucleic acid has been reduced or disabled, in other ways than mentioned above, for instance through exchanging capsid proteins, or fragments thereof, by capsid proteins, or fragments thereof, from other serotypes for which humans do not have, or have low levels of, neutralizing antibodies. Another example of this is the exchange of genes encoding capsid proteins with the genes encoding for capsid proteins from other serotypes. Also capsid proteins, or fragments thereof, may be exchanged for other capsid proteins, or fragments thereof, for which individuals are not capable of, or have a low capacity of, raising an immune response against.
  • An adenovirus nucleic acid may be altered further or instead of one or more of the alterations mentioned above, by inactivating or deleting genes encoding adenovirus late proteins such as, but not limited to, hexon, penton, fiber and/or protein IX.
  • In a preferred embodiment of the invention all genes encoding adenovirus proteins are deleted from the adenovirus nucleic acid, turning the nucleic acid into a minimal adenovirus vector.
  • In another preferred embodiment of the invention, the adenovirus nucleic acid is an Ad/AAV chimeric vector, wherein at least the integration means of an adeno-associated virus (AAV) is incorporated into the adenovirus nucleic acid.
  • In a preferred embodiment, a vector or a nucleic acid, which may or may not be one and the same according to the invention, further comprises at least one non-adenovirus gene. Preferably, at least one of the non-adenovirus genes is selected from the group of genes encoding: an apolipoprotein, a ceNOS, a herpes simplex virus thymidine kinase, an interleukin-3, an interleukin-1α, an (anti) angiogenesis protein such as angiostatin, an anti-proliferation protein, a vascular endothelial growth factor (VGEF), a basic fibroblast growth factor (bFGF), a hypoxia inducible factor 1α (HIF-1α), a PAI-1 and a smooth muscle cell anti-migration protein.
  • In another aspect, the invention provides a cell for the production of a gene delivery vehicle provided with at least a tissue tropism for smooth muscle cells and/or endothelial cells. In another aspect, the invention provides a cell for the production of a gene delivery vehicle deprived of at least a tissue tropism for liver cells. In another aspect, the invention provides a cell for the production of a gene delivery vehicle provided with at least a tissue tropism for smooth muscle cells and/or endothelial cells and deprived of at least a tissue tropism for liver cells. In a preferred embodiment, the cell is an adenovirus packaging cell, wherein an adenovirus nucleic acid is packaged into an adenovirus capsid. In one aspect of an adenovirus packaging cell of the invention all proteins required for the replication and packaging of an adenovirus nucleic acid, except for the proteins encoded by early region 1, are provided by genes incorporated in the adenovirus nucleic acid. The early region 1 encoded proteins in this aspect of the invention may be encoded by genes incorporated into the cells genomic DNA. In a preferred embodiment of the invention said cell is PER.C6 (ECACG deposit number 96022940). In general, when gene products required for the replication and packaging of adenovirus nucleic acid into adenovirus capsid are not provided by an adenovirus nucleic acid, they are provided by the packaging cell, either by transient transfection, or through stable transformation of said packaging cell. However, a gene product provided by the packaging cell may also be provided by a gene present on said adenovirus nucleic acid. For instance, fiber protein may be provided by the packaging cell, for instance through transient transfection, and may be encoded by the adenovirus nucleic acid. This feature can among others be used to generate adenovirus capsids comprising of fiber proteins from two different viruses.
  • The gene delivery vehicles of the invention are useful for the treatment of cardiovascular disease or disease treatable by nucleic acid delivery to endothelial cells or smooth muscle cells. A non-limiting example of the latter is for instance cancer, where the nucleic acid transferred comprises a gene encoding an anti-angiogenesis protein.
  • The gene delivery vehicles of the invention may be used as a pharmaceutical for the treatment of diseases. Alternatively, gene delivery vehicles of the invention may be used for the preparation of a medicament for the treatment of diseases.
  • In one aspect, the invention provides an adenovirus capsid with or provided with a tissue tropism for smooth muscle cells and/or endothelial cells wherein the capsid preferably comprises proteins from at least two different adenoviruses and wherein at least a tissue tropism-determining fragment of a fiber protein is derived from a subgroup B adenovirus, preferably of adenovirus 16. In another aspect, the invention provides an adenovirus capsid deprived of a tissue tropism for liver cells wherein the capsid preferably comprises proteins from at least two different adenoviruses and wherein at least a tissue tropism-determining fragment of a fiber protein is derived from a subgroup B adenovirus, preferably of adenovirus 16.
  • In one embodiment, the invention comprises the use of an adenovirus capsid, for the delivery of nucleic acid to smooth muscle cells and/or endothelial cells. In another embodiment, the invention comprises the use of an adenovirus capsid for preventing the delivery of nucleic acid to liver cells.
  • The adenovirus capsids of the invention may be used for the treatment of cardiovascular disease or a disease treatable by nucleic acid delivery to endothelial cells or smooth muscle cells. An example of the latter is, for instance, cancer where the nucleic acid transferred comprises a gene encoding an anti-angiogenesis protein.
  • The adenovirus capsids of the invention may be used as a pharmaceutical for the treatment of diseases. Alternatively, adenovirus capsids of the invention may be used for the preparation of a medicament for the treatment of diseases.
  • In another aspect of the invention, construct pBr/Ad.BamRΔfib (ECACC deposit number 01121708, deposited on Dec. 12, 2001 with the Centre for Applied Microbiology and Research Authority (European Collection of Animal Cell Cultures), Porton Down, Salisbury, Wiltshire SP4, OJG, United Kingdom, an International Depository Authority, in accordance with the Budapest Treaty is provided, comprising adenovirus 5 sequences 21562-31094 and 32794-35938.
  • In another aspect of the invention, construct pBr/AdBamRfib16 (ECACC deposit number 01121710, deposited on Dec. 12, 2001 with the Centre for Applied Microbiology and Research Authority (European Collection of Animal Cell Cultures), Porton Down, Salisbury, Wiltshire SP4, OJG, United Kingdom, an International Depository Authority, in accordance with the Budapest Treaty is provided, comprising adenovirus 5 sequences 21562-31094 and 32794-3598, further comprising an adenovirus 16 gene encoding fiber protein.
  • In yet another aspect of the invention, construct pBr/AdBamR.pac/fib16 (ECACC deposit number 01121709, deposited on Dec. 12, 2001 with the Centre for Applied Microbiology and Research Authority (European Collection of Animal Cell Cultures), Porton Down, Salisbury, Wiltshire SP4, OJG, United Kingdom, an International Depository Authority, in accordance with the Budapest Treaty, comprising adenovirus 5 sequences 21562-31094 and 32794-35938 is provided, comprising an adenovirus 16 gene encoding fiber protein, and further comprising a unique PacI-site in the proximity of the adenovirus 5 right terminal repeat, in the non-adenovirus sequence backbone of the construct.
  • In another aspect of the invention, construct pWE/Ad.AflIIrITRfib16 (ECACC deposit number 01121711, deposited on Dec. 12, 2001 with the Centre for Applied Microbiology and Research Authority (European Collection of Animal Cell Cultures), Porton Down, Salisbury, Wiltshire SP4, OJG, United Kingdom, an International Depository Authority is provided, in accordance with the Budapest Treaty comprising Ad5 sequence 3534-31094 and 32794-35938, further comprising an adenovirus 16 gene encoding fiber protein.
  • In another aspect of the invention, construct pWE/Ad.AflIIrITRDE2Afib16 (ECACC deposit number 01121712, deposited on Dec. 12, 2001 with the Centre for Applied Microbiology and Research Authority (European Collection of Animal Cell Cultures), Porton Down, Salisbury, Wiltshire SP4, OJG, United Kingdom, an International Depository Authority, in accordance with the Budapest Treaty, is provided comprising Ad5 sequences 3534-22443 and 24033-31094 and 32794-35938, further comprising an adenovirus 16 gene encoding fiber protein.
  • In the numbering of the sequences mentioned above, the number is depicted until and not until plus.
  • In a preferred embodiment, the constructs are used for the generation of a gene delivery vehicle or an adenovirus capsid with a tissue tropism for smooth muscle cells and/or endothelial cells.
  • In another aspect, the invention provides a library of adenovirus vectors, or gene delivery vehicles which may be one and the same or not, comprising a large selection of non-adenovirus nucleic acids. In another aspect of the invention, adenovirus genes encoding capsid proteins are used to generate a library of adenovirus capsids comprising proteins derived from at least two different adenoviruses, the adenoviruses preferably being derived from two different serotypes, wherein preferably one serotype is an adenovirus of subgroup B. In a particularly preferred embodiment, a library of adenovirus capsids is generated comprising proteins from at least two different adenoviruses and wherein at least a tissue tropism-determining fragment of fiber protein is derived from an adenovirus of subgroup B, preferably of adenovirus 16.
  • A fiber protein of adenovirus 16 preferably comprises the sequence given in FIG. 9. However, within the scope of the present invention analogous sequences may be obtained by using codon degeneracy. Alternatively, amino-acid substitutions or insertions or deletions may be performed as long as the tissue tropism-determining property is not significantly altered. Such amino-acid substitutions may be within the same polarity group or without.
  • In the following, the invention is illustrated by a number of non-limiting examples.
  • EXAMPLES Example 1 Generation of Adenovirus Serotype 5 Based Viruses with Chimeric Fiber Proteins
  • Generation of Adenovirus Template Clones Lacking DNA Encoding for Fiber
  • The fiber coding sequence of adenovirus serotype 5 is located between nucleotides 31042 and 32787. To remove the adenovirus serotype 5 DNA encoding fiber we started with construct pBr/Ad.Bam-rITR (FIG. 1; ECACC deposit P97082122). From this construct a NdeI site was first removed. For this purpose, pBr322 plasmid DNA was digested with NdeI after which protruding ends were filled using Klenow enzyme. This pBr322 plasmid was then re-ligated, digested with NdeI and transformed into E. coli DH5α. The obtained pBr/ΔNdeI plasmid was digested with ScaI and SalI and the resulting 3198 bp vector fragment was ligated to the 15349 bp ScaI-SalI fragment derived from pBr/Ad.Bam-rITR, resulting in plasmid pBr/Ad.Bam-rITRΔNdeI which hence contained a unique NdeI site. Next a PCR was performed with oligonucleotides “NY-up” and “NY-down” (FIG. 2). During amplification, both a NdeI and a NsiI restriction site were introduced to facilitate cloning of the amplified fiber DNAs. Amplification consisted of 25 cycles of each for 45 seconds at 94° C., 1 minute at 60° C., and 45 seconds at 72° C. The PCR reaction contained 25 pmol of oligonucleotides NY-up or NY-down, 2 mM dNTP, PCR buffer with 1.5 mM MgCl2, and 1 unit of Elongase heat stable polymerase (Gibco, The Netherlands). One-tenth of the PCR product was run on an agarose gel which demonstrated that the expected DNA fragment of ±2200 bp was amplified. This PCR fragment was subsequently purified using Geneclean kit system (Bio101 Inc.). Then, both the construct pBr/Ad.Bam-rITRΔNdeI as well as the PCR product were digested with restriction enzymes NdeI and SbfI. The PCR fragment was subsequently cloned using T4 ligase enzyme into the NdeI and Sbf I sites thus generating pBr/Ad.BamRΔFib (FIG. 3).
  • Amplification of Fiber Sequences from Adenovirus Serotypes:
  • To enable amplification of the DNAs encoding fiber protein derived from alternative serotypes degenerate oligonucleotides were synthesized. For this purpose, first known DNA sequences encoding for fiber protein of alternative serotypes were aligned to identify conserved regions in both the tail region as well as the knob region of the fiber protein. From the alignment, which contained the nucleotide sequence of 19 different serotypes representing all 6 subgroups, (degenerate) oligonucleotides were synthesized (see Table I, SEQ ID NOS:1-13). Also shown in Table 3 is the combination of oligonucleotides used to amplify the DNA encoding fiber protein of a specific serotype. The amplification reaction (50 μl) contained 2 mM dNTPs, 25 pmol of each oligonucleotide, standard 1×PCR buffer, 1.5 mM MgCl2, and 1 Unit Pwo heat stable polymerase (Boehringer Mannheim) per reaction. The cycler program contained 20 cycles, each consisting of 30 seconds at 94° C., 60 seconds at 60-64° C., and 120 seconds at 72° C. One-tenth of the PCR product was run on an agarose gel to demonstrate that a DNA fragment was amplified. From each different template, two independent PCR reactions were performed.
  • Generation of Chimeric Adenoviral DNA Constructs:
  • All amplified fiber DNAs as well as the vector (pBr/Ad.BamRΔFib) (ECACC deposit number 01121708) were digested with NdeI and NsiI. The digested DNAs were subsequently run on an agarose gel after which the fragments were isolated from the gel and purified using the Geneclean kit (Bio101 Inc). The PCR fragments were then cloned into the NdeI and NsiI sites of pBr/AdBamRΔFib (ECACC deposit number 01121708), thus generating pBr/AdBamRFibXx (where XX stands for the serotype number of which the fiber DNA was isolated). The inserts generated by PCR were sequenced to confirm correct amplification. The obtained sequences of the different fiber genes are shown in FIG. 4.
  • Generation of Recombinant Adenovirus Chimeric for Fiber Protein:
  • To enable efficient generation of chimeric viruses an AvrII fragment from the pBr/AdBamRFib16 (ECACC deposit number 01121710), pBr/AdBamRFib28, pBr/AdBamRFib40-L constructs was subcloned into the vector pBr/Ad.Bam-rITR.pac#8 (ECACC deposit #P97082121) replacing the corresponding sequences in this vector. pBr/Ad.Bam-rITR.pac#8 has the same adenoviral insert as pBr/Ad.Bam-rITR but has a PacI site near the rITR that enables the ITR to be separated from the vector sequences. The constuct pWE/Ad.AflII-Eco was generated as follows. PWE.pac was digested with ClaI and the 5 prime protruding ends were filled in with Klenow enzyme. The DNA was then digested with PacI and isolated from agarose gel. PWE/AflIIrITR was digested with EcoRI and after treatment with Klenow enzyme digested with PacI. The large 24 kb fragment containing the adenoviral sequences was isolated from agarose gel and ligated to the ClaI digested and blunted pWE.Pac vector. Use was made of the ligation express kit from Clontech. After transformation of XL10-gold cells from Stratagene, clones were identified that contained the expected construct. PWE/Ad.AlfII-Eco contains Ad5 sequences from basepairs 3534-27336. Three constructs, pClipsal-Luc (FIG. 5) digested with SalI, pWE/Ad.AflII-Eco digested with PacI and EcoRI and pBr/AdBamR.pac/fibXX digested with BamHI and PacI were transfected into adenovirus producer cells (PER.C6, Fallaux et al., 1998). FIG. 6 schematically depicts the method and fragments used to generate the chimeric viruses. Only pBr/Ad.BamRfib12 was used without subcloning in the PacI containing vector and therefor was not liberated from vector sequences using PacI but was digested with ClaI which leaves approximately 160 bp of vector sequences attached to the right ITR. Furthermore, the pBr/Ad.BamRfib12 and pBr/Ad.BamRfib28 contain an internal BamHI site in the fiber sequences and were therefor digested with SalI which cuts in the vector sequences flanking the BamHI site. For transfection, 2 μg of pCLIPsal-Luc, and 4 μg of both pWE/Ad.AflII-Eco and pBr/AdBamR.pac/fibXX were diluted in serum free DMEM to 100 μl total volume. To this DNA suspension, 100 μl 2.5× diluted lipofectamine (Gibco) in serum-free medium were added. After 30 minutes at room temperature, the DNA-lipofectamine complex solution was added to 2.5 ml of serum-free DMEM which was subsequently added to a T25 cm2 tissue culture flask. This flask contained PER.C6 cells that were seeded 24-hours prior to transfection at a density of 1×106 cells/flask. Two hours later, the DNA-lipofectamine complex containing medium was diluted once by the addition of 2.5 ml DMEM supplemented with 20% fetal calf serum. Again 24 hours later, the medium was replaced by fresh DMEM supplemented with 10% fetal calf serum. Cells were cultured for 6-8 days, subsequently harvested, and freeze/thawed 3 times. Cellular debris was removed by centrifugation for 5 minutes at 3000 rpm at room temperature. Of the supernatant (12.5 ml), 3-5 ml was used to infect again PER.C6 cells (T80 cm2 tissue culture flasks). This re-infection results in full cytopathogenic effect (CPE) after 5-6 days after which the adenovirus is harvested as described above.
  • Production of Fiber Chimeric Adenovirus:
  • 10 ml of the above described crude lysate was used to inoculate a 1 liter fermentor which contained 17-1.5×106 PER.C6 cells/ml growing in suspension. Three days after inoculation, the cells were harvested and pelleted by centrifuging for 10 minutes at 1750 rpm at room temperature. The chimeric adenovirus present in the pelleted cells was subsequently extracted and purified using the following downstream processing protocol. The pellet was dissolved in 50 ml 10 mM NaPO4 and frozen at −20° C. After thawing at 37° C., 5.6 ml deoxycholate (5% w/v) was added after which the solution was homogenated. The solution was subsequently incubated for 15 minutes at 37° C. to completely crack the cells. After homogenizing the solution, 1875 μl (1M) MgCl2 was added and 5 ml 100% glycerol. After the addition of 375 μl DNase (10 mg/ml) the solution was incubated for 30 minutes at 37° C. Cell debris was removed by centrifugation at 1880×g for 30 minutes at room temperature without the brake on. The supernatant was subsequently purified from proteins by loading on 10 ml of freon. Upon centrifugation for 15 minutes at 2000 rpm without brake at room temperature, three bands are visible of which the upper band represents the adenovirus. This band was isolated by pipetting after which it was loaded on a Tris/HCl (1M) buffered cesium chloride blockgradient (range: 1.2 to 1.4 g/ml). Upon centrifugation at 21000 rpm for 2.5 hours at 10° C., the virus was purified from remaining protein and cell debris since the virus in contrast to the other components, does not migrate into the 1.4 g/ml cesium chloride solution. The virus band is isolated after which a second purification using a Tris/HCl (1M) buffered continues gradient of 1.33 g/ml of cesium chloride is performed. After virus loading on top of this gradient, the virus is centrifuged for 17 hours at 55000 rpm at 10° C. Subsequently, the virus band is isolated and after the addition of 30 μl of sucrose (50 w/v) excess cesium chloride is removed by three rounds of dialysis, each round comprising 1 hour. For dialysis, the virus is transferred to dialysis slides (Slide-a-lizer, cut off 10000 kDa, Pierce, USA). The buffers used for dialysis are PBS which are supplemented with an increasing concentration of sucrose (round 1 to 3: 30 ml, 60 ml, and 150 ml sucrose (50% w/v)/1.5 liter PBS, all supplemented with 7.5 ml 2% (w/v) CaMgCl2). After dialysis, the virus is removed from the slide-a-lizer after which it is aliquoted in portions of 25 and 100 μl upon which the virus is stored at −85° C. To determine the number of virus particles per ml, 50 μl of the virus batch is run on a high pressure liquid chromatograph (HPLC) as described by Shamram et al. (1997). The virus titers were found to be in the same range as the Ad5.Luc virus batch (Ad5.Luc: 2.2×1011 vp/ml; Ad5.LucFib12: 1.3×1011 vp/ml; Ad5.LucFib16: 3.1×1012 vp/ml; Ad5.LucFib28: 5.4×1010 vp/ml; Ad5.LucFib40-L: 1.6×1012 vp/ml).
  • Example 2 Biodistribution of Chimeric Viruses after Intravenous Tail Vein Injection of Rats
  • To investigate the biodistribution of the chimeric adenoviruses carrying fiber 12, 16, 28, or 40-2, 1×1010 particles of each of the generated virus batches was diluted in 1 ml PBS after which the virus was injected in the tail vein of adult male Wag/Rij rats (3 rats/virus). As a control, Ad5 carrying the luciferase transgene was used. Forty-eight hours after the administration of the virus, the rats were sacrificed after which the liver, spleen, lung, kidney, heart, and brain were dissected. These organs were subsequently mixed with 1 ml of lysis buffer (1% Triton X-100/PBS) and minced for 30 seconds to obtain a protein lysate. The protein lysate was subsequently tested for the presence of transgene expression (luciferase activity) and the protein concentration was determined to express the luciferase activity per μg of protein. The results, Shown in Table II, demonstrate that in contrast to the Adenovirus serotype 5 control, none of the fiber chimeras are targeted specifically to the liver or to the spleen. This experiment shows that it is possible to circumvent the uptake of adenoviruses by the liver by making use of fibers of other serotypes. It also shows that the uptake by the liver is not correlated with the length of the fiber shaft, or determined solely by the ability of fiber knob to bind to CAR. The fibers used have different shaft lengths and, except for fiber 16, are derived from subgroups known to have a fiber that can bind CAR (Roelvink et al. 1998).
  • Example 3 Chimeric Viruses Display Differences in Endothelial and Smooth Muscle Cell Transduction
  • A) Infection of Human Endothelial Cells
  • Human endothelial cells (HUVEC) were isolated, cultured and characterized as described previously (Jaffe et al. 1973; Wijnberg et al. 1997). Briefly, cells were cultured on gelatin-coated dishes in M199 supplemented with 20 mM HEPES, pH 7.3 (Flow Lab., Irvine, Scotland), 10% (v/v) human serum (local blood bank), 10% (v/v) heat-inactivated newborn calf serum (NBCS) (GIBCO BRL, Gaithersburg, Md.), 150 μg/ml crude endothelial cell growth factor, 5 U/ml heparin (Leo Pharmaceutics Products, Weesp, The Netherlands), penicillin (100 IU/ml)/streptomycin (100 μg/ml) Boehringer Mannheim, Mannheim, Del.) at 37° C. under 5% (v/v) CO2/95% (v/v) air atmosphere. Cells used for experiments were between passage 1-3. In a first set of experiments 40000 HUVEC cells (a pool from 4 different individuals) were seeded in each well of 24-well plates in a total volume of 200 μl. Twenty-four hours after seeding, the cells were washed with PBS after which 200 μl of DMEM supplemented with 2% FGS was added to the cells. This medium contained various amounts of virus (MOI=50, 250, 1000, 2500, 5000, and 10,000). The viruses used were besides the control Ad5, the fiber chimeras 12, 16, 28, and 40-L (each infection in triplicate). Two hours after addition of the virus the medium was replaced by normal medium. Again forty-eight hours later cells were washed and lysed by the addition of 100 μl lysis buffer. In FIG. 7 a, results are shown on the transgene expression per microgram total protein after infection of HUVEC cells. These results show that fiber chimeras 12 and 28 are unable to infect HUVEC cells, that 40-L infects HUVECs with similar efficiency as the control Ad5 virus, and that fiber chimera 16 infects HUVECs significantly better. In a next set of experiments (n=8), the fiber 16 chimera was compared with the Ad5.Luc vector on HUVEC for luciferase activity after transduction with 2500 virus particles per cell of each virus. These experiments demonstrated that fiber 16 yields, on average, 8.1 fold increased luciferase activity (SD±4.6) as compared with Ad5. In a next experiment, an equal number of virus particles was added to wells of 24-well plates that contained different HUVEC cell concentrations. This experiment was performed since it is known that HUVECs are less efficiently infected with adenovirus serotype 5 when these cells reach confluency. For this purpose, HUVECs were seeded at 22,500, 45,000, 90,000, and 135,000 cells per well of 24-well-plates (in triplicate). Twenty-four hours later, these cells were infected as described above with medium containing 4.5×109 virus particles. The viruses used were besides the control adenovirus serotype 5, the chimera fiber 16. The result of the transgene expression (RLU) per microgram protein determined 48 hours after infection (see FIG. 7 b) shows that the fiber 16 chimeric adenovirus is also better suited to infect HUVEC cells even when these cells are 100% confluent which better mimics an in vivo situation. Since the Luciferase marker gene does not provide information concerning the number of cells infected another experiment was performed with adenovirus serotype 5 and the fiber 16 chimera, both carrying a green fluorescent protein (GFP) as a marker gene. This protein expression can be detected using a flow cytometer which renders information about the percentage of cells transduced as well as fluorescence per cell. In this experiment, cells were seeded at a concentration of 40,000 cells per well and were exposed to virus for 2 hours. The virus used was Ad5.GFP (8.4×1011 vp/ml) and Ad5.Fib16 GFP (5.1×1011 vp/ml). Cells were exposed to a virus concentration of 500 virus particles per cell. Flow cytometric analysis, 48 hours after virus exposure demonstrated that the fiber 16 virus gives higher transgene expression levels per cell since the median fluorescence, a parameter identifying the amount of GFP expression per cell, is higher with fiber 16 as compared to Ad5 (FIG. 7 c). These results are thus consistent and demonstrate that the fiber 16 chimeric virus is better suited to infect human primary endothelial cells as compared to Ad5.
  • B) Infection of Human Smooth Muscle Cells
  • Smooth muscle cells were isolated after isolation of EC (Weinberg et al. 1997). The veins were incubated with medium (DMEM) supplemented with penicillin/streptomycin) containing 0.075% (w/v) collagenase (Worthington Biochemical Corp., Freehold, N.J., USA). After 45 minutes, the incubation medium containing detached cells was flushed from the veins. Cells were washed and cultured on gelatin coated dishes in culture medium supplemented with 10% human serum at 37° C. under 5% (v/v) CO2/95% (v/v) air atmosphere. Cells used for experiments were between passage 3-6. We first tested the panel of chimeric fiber viruses versus the control adenovirus serotype 5 for the infection of human smooth muscle cells. For this purpose, 40000 human umbilical vein smooth muscle cells (HUVsmc) were seeded in wells of 24-well plates in a total volume of 200 μl. Twenty-four hours after seeding, the cells were washed with PBS after which 200 μl of DMEM supplemented with 2% FCS was added to the cells. This medium contained various amounts of virus (MOI=50, 250, 1250, 2500, and 5000). The viruses used were besides the control Ad5 the fiber chimeras 12, 16, 28 and 40-L (each infection in triplicate). Two hours after addition of the virus the medium was replaced by normal medium. Again forty-eight hours later, cells were washed and lysed by the addition of 100 μl lysis buffer. In FIG. 8 a, results are shown of the transgene expression per microgram total protein after infection of HUVsmc cells. These results show that fiber chimeras 12 and 28 are unable to infect HUVsmc cells, that 40-L infects HUVsmc with similar efficiency as the control Ad5 virus, and that fiber chimera 16 infects HUVsmc significantly better. In a next set of experiments, smooth muscle cells derived from saphenous vene, arteria Iliaca, left interior mammory artery (LIMA) and aorta were tested for infection with the fiber 16 chimera and Ad5 (both carrying luciferase as a marker gene). These experiments (n=11) demonstrated that, on average, the fiber 16 chimera yielded 61.4 fold increased levels in luciferase activity (SD±54.8) as compared to Ad5. The high standard deviation (SD) is obtained due to the finding that the adenoviruses used vary in their efficiency of infection of SMC derived from different human vessels. In a next experiment, an equal number of virus particles was added to wells of 24-well plates that contained different HUVsmc cell concentrations confluency. For this purpose, HUVsmc were seeded at 10,000, 20,000, 40,000, 60,000, and 80,000 cells per well of 24-well plates (in triplicate). Twenty-four hours later these cells were infected as described above with medium containing 2×108 virus particles. The viruses used were, besides the control adenovirus serotype 5, the chimera fiber 16. The result of the transgene expression (RLU) per microgram protein determined 48 hours after infection (see FIG. 8 b) shows that the fiber 16 chimeric adenovirus is better suited to infect smooth muscle cells even when these cells are 100% confluent which better mimics an in vivo situation.
  • To identify the number of SMCs transduced with the fiber 16 chimera and Ad5, we performed transduction experiments with Ad5.GFP and Ad5Fib16.GFP (identical batches as used for EC infections). Human umbilical vein SMC were seeded at a concentration of 60000 cells per well in 24-well plates and exposed for 2 hours to 500 or 5000 virus particles per cell of Ad5.GFP or Ad5Fib16.GFP. Forty-eight hours after exposure, cells were harvested and analyzed using a flow cytometer. The results obtained show that the fiber 16 mutant yields approximately 10 fold higher transduction of SMC since the GFP expression measured after transduction with 5000 virus particles of Ad5.GFP is equal to GFP expression after transduction with 500 virus particles per cell of the fiber 16 chimera (FIG. 8 c).
  • C) Subgroup B Fiber Mutants Other than Fiber 16
  • The shaft and knob of fiber 16 are derived from adenovirus serotype 16 which, as described earlier, belongs to subgroup B. Based on hemagglutination assays, DNA restriction patterns, and neutralization assays, the subgroup B viruses have been further subdivided into subgroup B1 and B2 (Wadell et al. 1984). Subgroup B1 members include serotypes 3, 7, 16, 21, and 51. Subgroup B2 members include 11, 14, 34, and 35. To test whether the increased infection of smooth muscle cells is a trade of all fibers derived from subgroup B or specific for one or more subgroup B fiber molecules, we compared fiber 16 and fiber 51 (both subgroup B1) with fiber 11 and fiber 35 (both subgroup B2). For this purpose, HUVsmc were infected with increasing amounts of virus particles per cell (156, 312, 625, 1250, 2500, 5000). The fiber mutants all carry the Luciferase marker gene (Ad5Fib11.Luc: 1.1×1012 vp/ml; Ad5Fib35Luc: 1.4×1012 vp/ml; Ad5Fib35Luc: 1.4×1012 vp/ml; Ad5Fib51Luc: 1.0×1012 vp/ml). Based on the Luciferase activity measured and shown in FIG. 8 d, efficient infection of SMC is not a general trade of all subgroup B fiber molecules. Clearly, fiber 16 and fiber 11 are better suited for infection of SMC than fiber 35 and fiber 51. Nevertheless, all subgroup B fiber mutants tested infect SMC better than Ad5.
  • D) Organ Culture Experiments
  • We next identified whether the observed difference in transduction of EC and SMC using the fiber 16 chimera or the Ad5 can also be demonstrated in organ culture experiments. Hereto, we focused on the following tissues: 1) Human Saphenous vein: the vein used in approximately 80% of all clinical vein grafting procedures. 2) Human pericard/epicard: for delivery of recombinant adenoviruses to the pericardial fluid which after infection of the pericardial or epicardial cells produce the protein of interest from the transgene carried by the adenovirus. 3) Human coronary arteries: percutaneous transluminal coronary angioplasty (PTCA) to prevent restenosis. Of the coronary arteries we focused on the left artery descending (LAD) and right coronary artery (RCA).
  • Parts of a human saphenous vein left over after vein graft surgery were sliced into pieces of approximately 0.5 cm. These pieces (n=3) were subsequently cultured for 2 hours in 200 ml of 5×1010 virus particles per ml. After two hours virus exposure, the pieces were washed with PBS and cultured for another 48 hours at 37° C. in a 10% CO2 incubator. The pieces were then washed, fixated and stained for LacZ transgene expression. The viruses were Ad5.LacZ (2.2×1012 vp/ml), the fiber 16 chimera: Ad5Fib16.LacZ (5.2×1011 vp/ml), and a fiber 51 chimera: Ad5Fib51.LacZ (2.1×1012 vp/ml). The pieces of saphenous vein were macroscopically photographed using a digital camera. Based on LacZ transgene expression obtained after 2 hours of virus exposure on saphenous vein slices, both the fiber 16 and the fiber 51 chimeric viruses give higher infection since much more blue staining is observed using these viruses as compared to Ad5.LacZ (FIG. 8 e). Identical experiments as described on saphenous vein were performed with human pericard and the human coronary arteries: RCA and LAD. Results of these experiments (FIGS. 8 f-8 g-8 h respectively) together with the experiments performed on primary cells confirmed the superiority of the fiber 16 and 51 mutants as compared to Ad5 in infecting human cardiovascular tissues.
  • E) CAR and Integrin Expression on Human EC and SMC
  • From the above described results, it is clear that the chimeric adenovirus with the shaft and knob from fiber 16 is well suited to infect endothelial cells and smooth muscle cells. Thus, by changing the fiber protein on Ad5 viruses we are able to increase infection of cells that are poorly infected by Ad5. The difference between Ad5 and Ad5Fib16, although significant on both cell types, is less striking on endothelial cells as compared to smooth muscle cells. This may reflect differences in receptor expression. For example, HUVsmc has significantly more αvβ5 integrins than HUVEC (see below). Alternatively, this difference may be due to differences in expression of the receptor of fiber 16. Ad5.LucFib16 infects umbilical vein smooth muscle cells approximately 8 fold better than umbilical vein endothelial cells whereas in case of Ad5.Luc viruses endothelial cells are better infected than smooth muscle cells. To test whether Ad5 infection correlated with receptor expression of these cells the presence of CAR and α-integrins was assayed on a flow cytometer. For this purpose 1×105 HUVEC cells or HUVsmc were washed once with PBS/0.5% BSA after which the cells were pelleted by centrifugation for 5 minutes at 1750 rpm at room temperature. Subsequently, 10 μl of a 100 times diluted αvβ3 antibody (Mab 1961, Brunswick Chemie, Amsterdam, The Netherlands), a 100 times diluted antibody αvB5 (antibody (Mab 1976, Brunswick Chemie, Amsterdam, The Netherlands), or 2000 times diluted CAR antibody was a kind gift of Dr. Bergelson, Harvard Medical School, Boston, Mass., USA (Hsu et al.) was added to the cell pellet after which the cells were incubated for 30 minutes at 4° C. in a dark environment. After this incubation, cells were washed twice with PBS/0.5% BSA and again pelleted by centrifugation for 5 minutes at 1750 rpm room temperature. To label the cells, 10 ml of rat anti-mouse IgG1 labeled with phycoerythrine (PE) was added to the cell pellet upon which the cells were again incubated for 30 minutes at 4° C. in a dark environment. Finally, the cells were washed twice with PBS/0.5% BSA and analyzed on a flow cytometer. The results of these experiments are shown in table III. From the results it can be concluded that HUVsmc do not express detectable levels of CAR confirming that these cells are difficult to transduce with an adenovirus which enters the cells via the CAR receptor.
  • F) Infection of Human A549 Cells
  • As a control for the experiments performed on endothelial cells and smooth muscle cells, A549 cells were infected to establish whether an equal amount of virus particles of the different chimeric adenoviruses show significant differences in transgene expression on cell lines that are easily infected by adenovirus. This is to investigate whether the observed differences in infection efficiency on endothelial and smooth muscle cells are cell type specific. For this purpose, 105 A549 cells were seeded in 24-well plates in a volume of 200 PI. Two hours after seeding, the medium was replaced by medium containing different amounts of particles of either fiber chimera 5, 12, 16, or 40-L (MOI=0, 5, 10, 25, 100, 500). Twenty-four hours after the addition of virus, the cells were washed once with PBS after which the cells were lysed by the addition of 100 μl lysis buffer to each well (1% Triton X-100 in PBS) after which transgene expression (Luciferase activity) and the protein concentration was determined. Subsequently, the luciferase activity per μg protein was calculated. The data, shown in Table IV, demonstrate that Ad5.Luc viruses infect A549 cells most efficient while the infection efficiency of Ad5LucFib16 or Ad5.LucFib40-L is a few times lower. This means that the efficient infection of endothelial cells and especially smooth muscle cells is due to differences in binding of the virus to these cells and not to the amount of virus or the quality of the viruses used.
    TABLE I
    Serotype Tail oligonucleotide Knob oligonucleotide
    4 A 1
    8 B 2
    9 B 2
    12 E 3
    16 C 4
    19p B 2
    28 B 2
    32 B 2
    36 B 2
    37 B 2
    40-1 D 5
    40-2 D 6
    41-3 D 5
    41-1 D 7
    49 B 2
    50 B 2
    51 C 8
    A: 5′ -CCC GTG TAT CCA TAT GAT GCA GAC AAC GAC CGA CC-3′ (SEQ ID NO:1)
    B: 5′ -CCC GTC TAC CCA TAT GGC TAC GCG CGG-3′ (SEQ ID NO:2)
    C: 5′ -CCK GTS TAC CCA TAT GAA GAT GAA AGC-3′ (SEQ ID NO:3)
    D: 5′ -CCC GTC TAC CCA TAT GAC ACC TYC TCA ACT C-3′ (SEQ ID NO:4)
    E 5′ -CCC GTT TAC CCA TAT GAC CCA TTT GAC ACA TCA GAC-3′ (SEQ ID NO:5)
    1: 5′ -CCG ATG CAT TTA TTG TTG GGC TAT ATA GGA-3′ (SEQ ID NO:6)
    2: 5′ -CCG ATG CAT TYA TTC TTG GGC RAT ATA GGA-3′ (SEQ ID NO:7)
    3: 5′ -CCG ATG CAT TTA TTC TTG GGR AAT GTA WGA AAA GGA-3′ (SEQ ID NO:8)
    4: 5′ -CCG ATG CAT TCA GTC ATC TTC TCT GAT ATA-3′ (SEQ ID NO:9)
    5: 5′ -CCG ATG CAT TTA TTG TTC AGT TAT GTA GCA-3′ (SEQ ID NO:10)
    6: 5′ -GCC ATG CAT TTA TTG TTC TGT TAC ATA AGA-3′ (SEQ ID NO:11)
    7: 5′ -CCG TTA ATT AAG CCC TTA TTG TTC TGT TAC ATA AGA A-3′ (SEQ ID NO:12)
    8: 5′ -CCG ATG CAT TCA GTC ATC YTC TWT AAT ATA-3′ (SEQ ID NO:13)
  • TABLE II
    Organ Control Ad5 Fib 12 Fib 16 Fib 28 Fib 40-L
    Liver 740045 458 8844 419 2033
    Spleen 105432 931 3442 592 16107
    Lung 428 315 334 316 424
    Kidney 254 142 190 209 224
    Heart 474 473 276 304 302
    Brain 291 318 294 323 257
  • TABLE III
    Cell line αVβ3 αVβ5 CAR
    HUVEC 70% 98.3% 18.9% 18.1%
    HUVEC 100% 97.2% 10.5%  7.2%
    HUVsmc 70% 95.5% 76.6%  0.3%
    HUVsmc 100% 92.2% 66.5%  0.3%
    PER.C6  7.8% 16.8% 99.6%
  • TABLE IV
    MOI (vp/Cell) Control Ad5 Fiber 12 Fiber 16 Fiber 40-L
    0 0 0 0 0
    5 1025 46 661 443
    10 1982 183 1704 843
    25 4840 200 3274 2614
    100 21875 1216 13432 11907
    500 203834 3296 93163 71433
  • REFERENCES
  • Arnberg, N., Mei Y. and Wadell G. (1997) Fiber genes of adenoviruses with tropism for the eye and the genital tract. Virology 227: 239-244.
    • Bergelson, J. M., Cunningham, J. A., Droguett, G., Kurt-Jones, E. A., Krithivas, A., Hong, J. S., Horwitz, M. S., Crowell, R. L. and Finberg, R. W. (1997) Isolation of a common receptor for coxsackie B virus and adenoviruses 2 and 5. Science 275: 1320-1323.
    • Bout, A. (1997) Gene therapy, p. 167-182. In: D. J. A. Crommelin and R. D. Sindelar (ed.), Pharmaceutical Biotechnology, Harwood Academic Publishers.
    • Bout, A. (1996) Prospects for human gene therapy. Eur. J. Drug Met. and Pharma., 2, 175-179.
    • Blaese, M., Blankenstein, T., Brenner, M., Cohen-Hagenauer, O. Gansbacher, B., Russel, S., Sorrentino, B. and Velu, T. (1995) Cancer Gene Ther. 2: 291-297
    • Brody, S. L. and Crystal, R. G. (1994) Adenovirus mediated in vivo gene transfer. Ann. N.Y. Acad. Sci. 716: 90-101.
    • Carter, A. J., Laird, J. R., Farb, A., Kufs, W., Wortham, D. C. and Virmani, R. (1994) Morphologic characteristics of lesion formation and time course of smooth muscle cell proliferation in a porcine proliferative restenosis model. J. Am. Coll. Cardiol. 24: 1398-1405.
    • Chroboczek J., Ruigrok R. W. H., and Cusack S. (1995) Adenovirus fiber, p. 163-200. In: W. Doerfler and P. Bohm (ed.), The molecular repertoire of adenoviruses, I. Springer-Verlag, Berlin.
    • Defer C., Belin M., Caillet-Boudin M. and Boulanger P. (1990) Human adenovirus-host cell interactions, comparative study with members of subgroup B and C. Journal of Virology 64 (8): 3661-3673.
    • Fallaux, F. J., Bout., A, van der Velde, I et al. (1998) New helper cells and matched E1-deleted adenovirus vectors prevent generation of replication competent adenovirus. Human Gene Therapy, 9, p 1909-1917.
    • Francki, R. I. B., Fauquet, C. M., Knudson, D. L., and Brown, F. (1991) Classification and nomenclature of viruses. Fifth report of the international committee on taxonomy of viruses. Arch, Virol. Suppl. 2: 140-144.
    • Gall J., Kass-Eisler A., Leinwand L., and Falck-Pedersen E. (1996) Adenovirus type 5 and 7 capsid chimera: fiber replacement alters receptor tropism without affecting primary immune neutralization epitopes. Journal of Virology 70 (4): 2116-2123.
    • Greber, U. F., Willets, M., Webster, P., and Helenius, A. (1993) Stepwise dismantling of adenovirus 2 during entry into cells. Cell 75: 477-486.
    • Hynes, R. O. (1992) Integrins: versatility, modulation and signaling in cell adhesion. Cell 69: 11-25.
    • Herz, J. and Gerard, R. D. (1993) Adenovirus-mediated transfer of low density lipoprotein receptor gene acutely accelerates cholesterol clearance in normal mice. Proc. Natl. Acad. Sci. U.S.A. 96: 2812-2816.
    • Hierholzer, J. C. (1992) Adenovirus in the immunocompromised host. Clin. Microbiol Rev. 5, 262-274.
    • Hierholzer, J. C., Wigand, R., Anderson, L. J., Adrian, T., and Gold, J. W. M. (1988) Adenoviruses from patients with AIDS: a plethora of serotypes and a description of five new serotypes of subgenus D (types 43-47). J. Infect. Dis. 158, 804-813.
    • Hong, S. S., Karayan, L., Tournier, J., Curiel, D. T. and Boulanger, P. A. (1997) Adenovirus type 5 fiber knob binds to MHC class I α2 domain at the surface of human epithelial and B lymphoblastoid cells. EMBO J. 16: 2294-2306.
    • Hsu, K. H., Lonberg-Holm, K., Alstein, B. and Crowell, R. L. (1988) A monoclonal antibody specific for the cellular receptor for the group B coxsackieviruses, J. Virol 62(5): 1647-1652.
    • Huard, J., Lochmuller, H., Acsadi, G., Jani, A., Massie, B. and Karpati, G. (1995) The route of administration is a major determinant of the transduction efficiency of rat tissues by adenoviral recombinants. Gene Ther. 2: 107-115.
    • Ishibashi, M. and Yasue, H. (1984) The adenoviruses, H. S. Ginsberg, ed., Plenum Press, London, New York. Chapter 12, 497-561.
    • Jaffe, E. A., Nachman, R. L., Becker, C. G., Minick, C. R. (1973) Culture of endothelial cells derived from umbilical veins. Identification by morphologic and immunologic criteria. J. Clin. Invest. 52, 2745-2756.
    • Kass-Eisler, A., Falck-Pederson, E., Elfenbein, D. H., Alvira, M., Buttrick, P. M., and Leinwand, L. A. (1994) The impact of developmental stage, route of administration and the immune system on adenovirus-mediated gene transfer. Gene Ther. 1: 395-402.
    • Khoo, S. H., Bailey, A. S., De Jong, J. C., and Mandal, B. K. (1995) Adenovirus infections in human immunodeficiency virus-positive patients: Clinical features and molecular epidemiology. J. Infect. Dis 172, 629-637.
    • Kidd, A. H., Chroboczek, J., Cusack, S., and Ruigrok, R. W. (1993) Adenovirus type 40 virions contain two distinct fibers. Virology 192, 73-84.
    • Krasnykh, V. N., Mikheeva G. V., Douglas, J. T. and Curiel, D. T. (1996) Generation of recombinant adenovirus vectors with modified fibers for altering viral tropism. J. Virol. 70(10): 6839-6846.
    • Krasnykh, V. N., Dmitriev, I., Mikheeva, G., Miller, C. R., Belousova, N. and Curiel, D. T. (1998) Characterization of an adenovirus vector containing a heterologous peptide epitope in the HI loop of the fiber knob. J. Virol. 72(3): 1844-1852.
    • Law, L., Chillon, M., Bosch, A., Armentano, D., Welsh, M. J. and Davidson, B. L. (1998) Infection of primary CNS cells by different adenoviral serotypes: Searching for a more efficient vector. Abstract 1st Annual Meeting American Society of Gene Therapy, Seattle, Wash.
    • Leppard, K. N. (1997) E4 gene function in adenovirus, adenovirus vector and adeno-associated virus infections. J. Gen. Virol. 78: 2131-2138.
    • Lloyd Jones, D. M. and Bloch, K. D. (1996) The vascular biology of nitric oxide and its role in atherogenesis. Annu. Rev. Med. 47: 365-375.
    • Morgan, C., Rozenkrantz, H. S., and Mednis, B. (1969) Structure and development of viruses as observed in the electron microscope. Entry and uncoating of adenovirus. J. Virol 4, 777-796.
    • Roelvink, P. W., Kovesdi, I. and Wickham T. J. (1996) Comparative analysis of adenovirus fiber-cell interaction: Adenovirus type 2 (Ad2) and Ad9 utilize the same cellular fiber receptor but use different binding strategies for attachment. J. Virol. 70: 7614-7621.
    • Roelvink, P. W., Lizonova, A., Lee, J. G. N., Li, Y., Bergelson, J. M., Finberg, R. W., Brough, D. E., Kovesdi, I. and Wickham, T. J. (1998) The coxsackie-adenovirus receptor protein can function as a cellular attachment protein for adenovirus serotypes from subgroups A, C, D, E, and F. J. Virol. 72: 7909-7915.
    • Rogers, B. E., Douglas, J. T., Ahlem, C., Buchsbaum, D. J., Frincke, J. and Curiel, D. T. (1997) Use of a novel cross-linking method to modify adenovirus tropism. Gene Ther. 4: 1387-1392.
    • Schulick, A. H., Vassalli, G., Dunn, P. F., Dong, G., Rade, J. J., Zamarron, C. and Dichek, D. A. (1997) Established immunity precludes adenovirus-mediated gene transfer in rat carotid arteries.
    • Schnurr, D. and Dondero, M. E. (1993) Two new candidate adenovirus serotypes. Intevirol. 36, 79-83.
    • Schwartz, R. S., Edwards, W. D., Huber, K. C., Antoniudes, L. C., Bailey, K. R., Camrud, A. R., Jorgenson, M. A. and Holmes, D. R. Jr. (1993) Coronary restenosis: Prospects for solution and new perspectives from a porcine model. Mayo Clin. Proc. 68: 54-62.
    • Shi, Y., Pieniek, M., Fard, A., O'Brien, J., Mannion, J. D. and Zalewski, A. (1996) Adventitial remodeling after coronary arterial injury. Circulation 93: 340-348.
    • Shabram, P. W., Giroux, D. D., Goudreau, A. M., Gregory, R. J., Horn, M. T., Huyghe, B. G., Liu, X., Nunnally, M. H., Sugarman, B. J. and Sutjipto, S. (1997) Analytical anion-exchange HPLC of recombinant type-5 adenoviral particles. Hum. Gene Ther. 8(4): 453-465.
    • Signas, G., Akusjarvi, G., and Petterson, U. (1985) Adenovirus 3 fiberpolypeptide gene: Complications for the structure of the fiber protein. J. Virol. 53, 672-678.
    • Stevenson, S. C., Rollence, M., White B., Weaver L. and McClelland, A. (1995) Human adenovirus serotypes 3 and 5 bind to two different cellular receptors via the fiber head domain. J. Virol 69(5): 2850-2857.
    • Stevenson, S. C., Rollence, M., Marshall-Neff, J. and McClelland, A. (1997) Selective targeting of human cells by a chimeric adenovirus vector containing a modified fiber protein. J. Virology 71(6): 4782-4790.
    • Stouten, P. W. F., Sander, C., Ruigrok, R. W. H., and Cusack, S. (1992) New triple helical model for the shaft of the adenovirus fiber. J. Mol. Biol. 226, 1073-1084.
    • Svensson, V. and Persson, R. (1984) Entry of adenovirus 2 into Hela cells. J. Virol. 51, 687-694.
    • Van der Vliet, P. C. (1995) Adenovirus DNA replication In: W. Doerfler and P. Bohm (eds.) The molecular repertoire of adenoviruses II. Springer-Verlag, Berlin.
    • Varga, M. J., Weibull, C., and Everitt, E. (1991) Infectious entry pathway of adenovirus type 2. J. Virol 65, 6061-6070.
    • Varenne, O., Pislaru, S., Gillijns, H., Van Pelt, N., Gerard, R. D., Zoldhelyi, P., Van de Werf, F., Collen, D. and Janssens, S. P. (1998) Local adenovirus-mediated transfer of human endothelial nitric oxide synthetase reduces luminal narrowing after coronary angioplasty in pigs. Circulation 98: 919-926.
    • Wadell, G. (1984) Molecular Epidemiology of human adenoviruses Curr. Top. Microbiol. Immunol. 110, 191-220.
    • Wickham, T. J, Carrion, M. E. and Kovesdi, I. (1995) Targeting of adenovirus penton base to new receptors through replacement of its RGD motif with other receptor-specific peptide motifs. Gene Therapy 2: 750-756.
    • Wickham, T. J., Segal, D. M., Roelvink, P. W., Carrion, M. E., Lizonova, A., Lee, G-M., and Kovesdi, I. (1996) Targeted adenovirus gene transfer to endothelial and smooth muscle cells by using bispecific antibodies. J. Virol. 70 (10), 6831-6838.
    • Wickham, T. J., Mathias, P., Cherish, D. A., and Nemerow, G. R. (1993) Integrins avb3 and avb5 promote adenovirus internalization but not virus attachment. Cell 73, 309-319.
    • Wijnberg, M. J., Quax, P. H. A., Nieuwenbroek, N. M. E., Verheijen, J. H. (1997) The migration of human smooth muscle cells in vitro is mediated by plasminogen activation and can be inhibited by alpha(2) macro globulin receptor associated protein. Thromb. and Haemostas. 78, 880-886.
    • Wold, W. S., Tollefson, A. E. and Hermiston, T. W. (1995) E3 transcription unit of adenovirus. In: W. Doerfler and P. Bohm (eds.). The molecular repertoire of adenoviruses I. Springer-Verlag, Berlin.
    • Zabner, J., Armentano, D., Chillon, M., Wadsworth, S. C., and Welsh, M. J. (1998) Type 17 fiber enhances gene transfer Abstract 1st Annual Meeting American Society of Gene Therapy, Seattle, Wash.

Claims (54)

1. A viral gene delivery vehicle comprising at least a tissue tropism for cells selected from the group consisting of smooth muscle cells, endothelial cells, or smooth muscle cells and epothelial cells.
2. The viral gene delivery vehicle of claim 1, wherein said gene delivery vehicle has been deprived of at least a tissue tropism for liver cells.
3. The viral gene delivery vehicle of claim 1, wherein said tissue tropism is provided by a virus capsid.
4. The viral gene delivery vehicle of claim 3, wherein said virus capsid comprises protein fragments from at least two different viruses.
5. The viral gene delivery vehicle of claim 4, wherein at least one of said viruses is an adenovirus.
6. The viral gene delivery vehicle of claim 4, wherein at least one of said viruses is an adenovirus of subgroup B.
7. The viral gene delivery vehicle of claim 4, wherein at least one of said protein fragments comprises a tissue tropism-determining fragment of a fiber protein derived from a subgroup B adenovirus.
8. The viral gene delivery vehicle of claim 6, wherein said subgroup B adenovirus is adenovirus 16.
9. The viral gene delivery vehicle of claim 6, wherein protein fragments not derived from an adenovirus of subgroup B are derived from an adenovirus of subgroup C.
10. The viral gene delivery vehicle of claim 1, further comprising a nucleic acid derived from an adenovirus.
11. The viral gene delivery vehicle of claim 1, further comprising a nucleic acid derived from at least two different adenoviruses.
12. The viral gene delivery vehicle of claim 10, wherein said nucleic acid comprises at least one sequence encoding a fiber protein comprising at least a tissue tropism-determining fragment of a subgroup B adenovirus fiber protein.
13. The viral gene delivery vehicle of claim 10, wherein said nucleic acid derived from adenovirus is modified such that the capacity of said adenovirus nucleic acid to replicate in a target cell has been reduced or disabled.
14. The viral gene delivery vehicle of claim 10, wherein said nucleic acid derived from an adenovirus is modified such that a host immune system's capacity to mount an immune response against adenoviral proteins encoded by adenoviral nucleic acid has been reduced or disabled.
15. The viral gene delivery vehicle of claim 1, further comprising a minimal adenovirus vector or an Ad/AAV chimeric vector.
16. The viral gene delivery vehicle of claim 1, further comprising at least one non-adenoviral nucleic acid.
17. The viral gene delivery vehicle of claim 16, wherein at least one of said non-adenoviral nucleic acids is a gene selected from the group of genes encoding a protein selected from the group consisting of: an apolipoprotein, a nitric oxide synthase, a herpes simplex virus thymidine kinase, an interleukin-3, an interleukin-1α, an (anti) angiogenesis protein, an anti-proliferation protein, a smooth muscle cell anti-migration protein, a vascular endothelial growth factor (VGEF), a basic fibroblast growth factor, a hypoxia inducible factor 1α (HIF-1α) and a PAI-1.
18. The viral gene delivery vehicle of claim 5, wherein at least one of said viruses is an adenovirus of subgroup B.
19. The viral gene delivery vehicle of claim 7, wherein said subgroup B adenovirus is adenovirus 16.
20. A recombinant adenovirus of a subgroup C origin having a reduced tissue tropism for liver cells as compared to the corresponding wild type adenovirus of subgroup C origin, said recombinant adenovirus comprising:
a capsid comprising a chimeric fiber protein, wherein a knob domain of said chimeric fiber protein is of an adenovirus origin selected from the group consisting of adenovirus 12, adenovirus 16, adenovirus 28 and adenovirus 40-L.
21. A pharmaceutical composition comprising:
the recombinant adenovirus of claim 20; and
a suitable vehicle.
22. The recombinant adenovirus of claim 20, wherein said reduced tissue tropism is provided by a virus capsid.
23. The recombinant adenovirus of claim 22, wherein said virus capsid comprises protein fragments from at least two different viruses.
24. The recombinant adenovirus of claim 23, wherein at least one of said at least two different viruses is an adenovirus.
25. The recombinant adenovirus of claim 24, wherein at least one of said protein fragments comprises a tissue tropism-determining fragment of a fiber protein of a subgroup B adenovirus origin.
26. A cell for producing a recombinant adenovirus having a tissue tropism for smooth muscle cells, said cell comprising:
means for assembling said recombinant adenovirus;
wherein said means includes at least one adenoviral nucleic acid encoding an adenoviral fiber protein having at least a tissue tropism-determining fragment of a subgroup B adenoviral fiber protein.
27. The cell of claim 26, wherein said cell is or is derived from a PER.C6 cell (ECACC deposit number 96022940).
28. An adenovirus capsid having a tissue tropism for smooth muscle cells and/or endothelial cells wherein said adenovirus capsid comprises proteins from at least two different adenoviruses and wherein at least a tissue tropism-determining fragment of a fiber protein is derived from a subgroup B adenovirus.
29. An adenovirus capsid having a reduced tropism for liver cells as compared to the corresponding wild type adenovirus capsid, said adenovirus capsid comprising:
a fiber protein of an adenovirus of subgroup C origin; and
wherein at least a knob domain of the fiber protein is of an adenovirus origin selected from the group consisting of adenovirus 12, adenovirus 16, adenovirus 28, and adenovirus 40-L.
30. A method of delivering nucleic acid to cells selected from the group of cells consisting of smooth muscle cells, endothelial cells and both smooth muscle and endothelial cells, said method comprising:
administering to said cells an adenovirus capsid comprising proteins from at least two different adenoviruses,
wherein at least a tissue tropism-determining fragment of a fiber protein is derived from a subgroup B adenovirus.
31. A recombinant adenovirus of a subgroup C origin having an increased tropism for smooth muscle cells when compared to an adenovirus of serotype 5 comprising:
a recombinant adenovirus capsid comprising a fiber protein, wherein at least a knob domain of the fiber protein is of an adenovirus of a subgroup B origin;
wherein said adenovirus of subgroup B origin is selected from the group consisting of adenovirus 11, adenovirus 16, adenovirus 35, and adenovirus 51.
32. The recombinant adenovirus of claim 31, wherein at least one of said at least two different viruses is an adenovirus.
33. The recombinant adenovirus of claim 31, wherein said subgroup B adenovirus is adenovirus 16.
34. The recombinant adenovirus of claim 31, wherein at least one of said peptides is of adenovirus subgroup C origin.
35. The recombinant adenovirus of claim 31, further comprising an adenoviral nucleic acid incorporated within a genome of said recombinant adenovirus.
36. The recombinant adenovirus of claim 35, wherein said adenoviral nucleic acid comprises sequences from at least two different adenoviruses.
37. The recombinant adenovirus of claim 35, wherein said adenoviral nucleic acid comprises at least one sequence encoding a fiber protein having a tissue tropism-determining fragment of a subgroup B adenovirus fiber protein.
38. The recombinant adenovirus of claim 35, wherein said adenoviral nucleic acid is modified such that the capacity of said adenoviral nucleic acid to replicate in a target cell has been reduced or disabled.
39. The recombinant adenovirus of claim 31, wherein said recombinant adenovirus is a minimal adenovirus vector or an Ad/AAV chimeric vector.
40. The recombinant adenovirus of claim 31, further comprising at least one non-adenoviral nucleic acid incorporated within a genome of said recombinant adenovirus.
41. The recombinant adenovirus of claim 40, wherein at least one of said non-adenoviral nucleic acids is a gene encoding a protein selected from the group of proteins consisting of: an apolipoprotein, a nitric oxide synthase, a herpes simplex virus thymidine kinase, an interleukin-3, an interleukin-1α, an angiogenesis protein, an anti-angiogenesis protein, an anti-proliferation protein, a smooth muscle cell anti-migration protein, a vascular endothelial growth factor, a basic fibroblast growth factor, a hypoxia inducible factor 1α and a PAI-1.
42. A recombinant adenovirus capsid having an increased tropism for smooth muscle cells as compared to the corresponding wild type adenovirus, said recombinant adenovirus capsid comprising:
a chimeric fiber protein, wherein at least a knob domain of the chimeric fiber protein is of an adenovirus of subgroup B origin; and
wherein the adenovirus of subgroup B origin is selected from the group consisting of adenovirus 11, adenovirus 16, adenovirus 35, and adenovirus 51;
wherein a remaining part of the chimeric fiber protein is of an adenovirus of a subgroup C origin.
43. A recombinant adenovirus having a capsid with an increased tropism for smooth muscle cells as compared to the corresponding wild type adenovirus, said recombinant adenovirus comprising:
a chimeric fiber protein comprising at least the knob domain of a fiber protein of an adenovirus selected from the group consisting of adenovirus 11, adenovirus 16, adenovirus 35, and adenovirus 51;
wherein a remaining part of the chimeric fiber protein is of an adenovirus of a subgroup C origin.
44. The recombinant adenovirus of claim 43, further comprising an adenoviral nucleic acid incorporated within a genome of said recombinant adenovirus.
45. The recombinant adenovirus of claim 44, wherein said adenoviral nucleic acid comprises a sequence encoding the chimeric fiber protein.
46. The recombinant adenovirus of claim 43, wherein said recombinant adenovirus is a minimal adenovirus or an Ad/AAV chimeric vector.
47. The recombinant adenovirus of claim 43, wherein said different adenovirus serotype is an adenovirus serotype of subgroup C.
48. The recombinant adenovirus of claim 43, wherein said adenovirus of subgroup C origin is adenovirus serotype 5.
49. A cell for producing a recombinant adenovirus having a tissue tropism for smooth muscle cells, said cell comprising:
means for assembling the recombinant adenovirus;
wherein said means comprises at least one adenoviral nucleic acid encoding a chimeric adenoviral fiber protein having at least a knob domain of a fiber protein of adenovirus serotype 16; and wherein the remaining part of the fiber protein is of a different adenovirus serotype; and
wherein said cell is of a PER.C6 cell (ECACC deposit number 96022940) origin.
50. The cell of claim 49, wherein said different adenovirus serotype is an adenovirus of subgoup C.
51. The cell of claim 50, wherein said adenovirus of subgroup C is adenovirus serotype 5.
52. A recombinant adenovirus capsid having a reduced tropism for liver cells as compared to the corresponding wild type adenovirus, said recombinant adenovirus comprising:
a chimeric fiber protein comprising at least the knob domain of a fiber protein of adenovirus serotype 16;
wherein the remaining part of the fiber protein is of an adenovirus of a subgroup C origin.
53. The adenovirus capsid of claim 52, wherein said different adenovirus serotype is an adenovirus serotype of subgroup C.
54. The adenovirus capsid of claim 52, wherein said adenovirus of subgroup C origin is adenovirus serotype 5.
US11/018,669 1998-11-20 2004-12-20 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells Abandoned US20050169891A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/018,669 US20050169891A1 (en) 1998-11-20 2004-12-20 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US12/455,086 US7968087B2 (en) 1998-11-20 2009-05-28 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP98203921.6 1998-11-20
EP98203921 1998-11-20
US09/348,354 US20030017138A1 (en) 1998-07-08 1999-07-07 Chimeric adenoviruses
US09/444,284 US6929946B1 (en) 1998-11-20 1999-11-19 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US11/018,669 US20050169891A1 (en) 1998-11-20 2004-12-20 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/444,284 Continuation US6929946B1 (en) 1998-11-20 1999-11-19 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/455,086 Continuation US7968087B2 (en) 1998-11-20 2009-05-28 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells

Publications (1)

Publication Number Publication Date
US20050169891A1 true US20050169891A1 (en) 2005-08-04

Family

ID=34828606

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/444,284 Expired - Lifetime US6929946B1 (en) 1998-11-20 1999-11-19 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US11/018,669 Abandoned US20050169891A1 (en) 1998-11-20 2004-12-20 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US12/455,086 Expired - Fee Related US7968087B2 (en) 1998-11-20 2009-05-28 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/444,284 Expired - Lifetime US6929946B1 (en) 1998-11-20 1999-11-19 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/455,086 Expired - Fee Related US7968087B2 (en) 1998-11-20 2009-05-28 Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells

Country Status (1)

Country Link
US (3) US6929946B1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090110695A1 (en) * 2006-03-27 2009-04-30 Menzo Jans Emko Havenga Compositions Comprising a Recombinant Adenovirus and an Adjuvant
US11077156B2 (en) 2013-03-14 2021-08-03 Salk Institute For Biological Studies Oncolytic adenovirus compositions
US11130968B2 (en) 2016-02-23 2021-09-28 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
US11401529B2 (en) 2016-02-23 2022-08-02 Salk Institute For Biological Studies Exogenous gene expression in recombinant adenovirus for minimal impact on viral kinetics
US11813337B2 (en) 2016-12-12 2023-11-14 Salk Institute For Biological Studies Tumor-targeting synthetic adenoviruses and uses thereof

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE278794T1 (en) 1995-06-15 2004-10-15 Crucell Holland Bv PACKAGING SYSTEMS FOR HUMAN, HUMAN ADENOVIRUSES, FOR USE IN GENE THERAPY
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
US8236561B2 (en) * 1999-04-15 2012-08-07 Crucell Holland B.V. Efficient production of IgA in recombinant mammalian cells
US6492169B1 (en) 1999-05-18 2002-12-10 Crucell Holland, B.V. Complementing cell lines
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
ATE542904T1 (en) * 2001-10-29 2012-02-15 Crucell Holland Bv METHOD AND MEANS FOR PRODUCING PROTEINS WITH PREDEFINED POST-TRANSLATIONAL MODIFICATIONS
CA2857087C (en) 2011-11-28 2021-05-25 Crucell Holland B.V. Influenza virus vaccines and uses thereof

Citations (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4487829A (en) * 1982-03-23 1984-12-11 Massachusetts Institute Of Technology Production and use of monoclonal antibodies against adenoviruses
US4517686A (en) * 1982-08-04 1985-05-21 La Jolla Cancer Research Foundation Polypeptide
US4578079A (en) * 1982-08-04 1986-03-25 La Jolla Cancer Research Foundation Tetrapeptide
US4589881A (en) * 1982-08-04 1986-05-20 La Jolla Cancer Research Foundation Polypeptide
US4593002A (en) * 1982-01-11 1986-06-03 Salk Institute Biotechnology/Industrial Associates, Inc. Viruses with recombinant surface proteins
US4792525A (en) * 1982-08-04 1988-12-20 La Jolla Cancer Research Foundation Tetrapeptide
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
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
US5096815A (en) * 1989-01-06 1992-03-17 Protein Engineering Corporation Generation and selection of novel dna-binding proteins and polypeptides
US5166320A (en) * 1987-04-22 1992-11-24 University Of Connecticut Carrier system and method for the introduction of genes into mammalian cells
US5198346A (en) * 1989-01-06 1993-03-30 Protein Engineering Corp. Generation and selection of novel DNA-binding proteins and polypeptides
US5204445A (en) * 1988-10-03 1993-04-20 The Scripps Research Institute Peptides and antibodies that inhibit integrin-ligand binding
US5223394A (en) * 1989-04-10 1993-06-29 Biogen, Inc. Recombinant dna molecule comprising lymphocyte function-associated antigen 3 phosphatidylinositol linkage signal sequence
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5240846A (en) * 1989-08-22 1993-08-31 The Regents Of The University Of Michigan Gene therapy vector for cystic fibrosis
US5246921A (en) * 1990-06-26 1993-09-21 The Wistar Institute Of Anatomy And Biology Method for treating leukemias
US5332567A (en) * 1989-08-24 1994-07-26 Immunomedics Detection and treatment of infections with immunoconjugates
US5349053A (en) * 1990-06-01 1994-09-20 Protein Design Labs, Inc. Chimeric ligand/immunoglobulin molecules and their uses
US5436146A (en) * 1989-09-07 1995-07-25 The Trustees Of Princeton University Helper-free stocks of recombinant adeno-associated virus vectors
US5443953A (en) * 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
US5474935A (en) * 1990-05-23 1995-12-12 The United States Of America As Represented By The Department Of Health And Human Services Adeno-associated virus (AAV)-based eucaryotic vectors
US5521291A (en) * 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
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
US5543328A (en) * 1993-08-13 1996-08-06 Genetic Therapy, Inc. Adenoviruses having modified fiber proteins
US5547932A (en) * 1991-09-30 1996-08-20 Boehringer Ingelheim International Gmbh Composition for introducing nucleic acid complexes into higher eucaryotic cells
US5552311A (en) * 1993-09-14 1996-09-03 University Of Alabama At Birmingham Research Foundation Purine nucleoside phosphorylase gene therapy for human malignancy
US5559099A (en) * 1994-09-08 1996-09-24 Genvec, Inc. Penton base protein and methods of using same
US5585254A (en) * 1987-08-21 1996-12-17 University Of Colorado Foundation, Inc. Autonomous parvovirus gene delivery vehicles and expression 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
US5770442A (en) * 1995-02-21 1998-06-23 Cornell Research Foundation, Inc. Chimeric adenoviral fiber protein and methods of using same
US5861290A (en) * 1989-01-23 1999-01-19 Goldsmith; Mark A. Methods and polynucleotide constructs for treating host cells for infection or hyperproliferative disorders
US5922315A (en) * 1997-01-24 1999-07-13 Genetic Therapy, Inc. Adenoviruses having altered hexon proteins
US6127525A (en) * 1995-02-21 2000-10-03 Cornell Research Foundation, Inc. Chimeric adenoviral coat protein and methods of using same
US6329190B1 (en) * 1995-11-28 2001-12-11 Genvec, Inc. Targetting adenovirus with use of constrained peptide motifs
US6455314B1 (en) * 1998-09-11 2002-09-24 Genvec, Inc. Alternatively targeted adenovirus
US6878549B1 (en) * 1998-04-24 2005-04-12 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy

Family Cites Families (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2602790B1 (en) 1986-08-13 1990-06-01 Transgene Sa EXPRESSION OF A SPECIFIC TUMOR ANTIGEN BY A RECOMBINANT VECTOR VIRUS AND USE THEREOF FOR THE PREVENTIVE 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
JPH027631A (en) 1988-06-24 1990-01-11 Nec Corp Time division multiple access satellite communication system
ATE144793T1 (en) 1989-06-29 1996-11-15 Medarex Inc BISPECIFIC REAGENTS FOR AIDS THERAPY
CA2069960C (en) 1989-10-20 2004-03-02 Michael W. Fanger Bispecific heteroantibodies with dual effector functions
DE69033181T2 (en) 1989-10-20 1999-10-21 Dartmouth College IGA RECEPTOR-SPECIFIC MONOCLONAL ANTIBODIES
GB2246779B (en) 1990-08-03 1994-08-17 Delta Biotechnology Ltd Tumour-associated protease inhibitors targeted to tumour cells
GB9101550D0 (en) 1991-01-24 1991-03-06 Mastico Robert A Antigen-presenting chimaeric protein
JP3534749B2 (en) 1991-08-20 2004-06-07 アメリカ合衆国 Adenovirus-mediated gene transfer into the gastrointestinal tract
US6743623B2 (en) 1991-09-27 2004-06-01 Centre National De La Recherche Scientifique Viral recombinant vectors for expression in muscle cells
FR2681786A1 (en) 1991-09-27 1993-04-02 Centre Nat Rech Scient RECOMBINANT VECTORS OF VIRAL ORIGIN, PROCESS FOR OBTAINING SAME AND THEIR USE FOR THE EXPRESSION OF POLYPEPTIDES IN MUSCLE CELLS.
IL103059A0 (en) 1991-09-30 1993-02-21 Boehringer Ingelheim Int Conjugates for introducing nucleic acid into higher eucaryotic cells
AU692423B2 (en) 1992-09-25 1998-06-11 Institut National De La Sante Et De La Recherche Medicale Adenovirus vectors for the transfer of foreign genes into cells of the central nervous system, particularly in brain
GB9223084D0 (en) 1992-11-04 1992-12-16 Imp Cancer Res Tech Compounds to target cells
EP0668913B1 (en) 1992-11-18 2002-01-30 Arch Development Corporation Adenovirus-mediated gene transfer to cardiac and vascular smooth muscle
GB9300686D0 (en) 1993-01-15 1993-03-03 Imp Cancer Res Tech Compounds for targeting
AU6133394A (en) 1993-02-09 1994-08-29 Scripps Research Institute, The Targeting and delivery of genes and antiviral agents into cells by the adenovirus penton
DE4311651A1 (en) 1993-04-08 1994-10-13 Boehringer Ingelheim Int Virus for the transport of foreign DNA into higher eukaryotic cells
ES2180579T3 (en) 1993-05-10 2003-02-16 Univ Michigan TRANSFER OF GENES IN PANCREATIC EPITHELIAL CELLS.
US5631236A (en) * 1993-08-26 1997-05-20 Baylor College Of Medicine Gene therapy for solid tumors, using a DNA sequence encoding HSV-Tk or VZV-Tk
EP0716711A1 (en) 1993-09-03 1996-06-19 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Vector for gene therapy of the liver
FR2712812B1 (en) 1993-11-23 1996-02-09 Centre Nat Rech Scient Composition for the production of therapeutic products in vivo.
IT1271461B (en) 1993-12-01 1997-05-28 Menarini Ricerche Sud Spa ANTI-CD3 / ANTI-EGFR MONOCLONAL ANTI-BODY ANTIBODY, PROCESS FOR PRODUCTION AND ITS USE.
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
US5928944A (en) 1994-02-04 1999-07-27 The United States Of America As Represented By The Department Of Health And Human Services Method of adenoviral-medicated cell transfection
US6312699B1 (en) 1994-03-28 2001-11-06 Uab Research Foundation Ligands added to adenovirus fiber
US7252989B1 (en) 1994-04-04 2007-08-07 Board Of Regents, The University Of Texas System Adenovirus supervector system
DE69520496T2 (en) 1994-05-13 2001-07-12 Chiron Corp METHOD AND COMPOSITIONS AS VEHICLES FOR TARGETED INTRODUCTION OF GENES
US5571531A (en) 1994-05-18 1996-11-05 Mcmaster University Microparticle delivery system with a functionalized silicone bonded to the matrix
FR2721943B1 (en) 1994-06-29 1996-08-02 Rhone Poulenc Rorer Sa ADENOVIRUS COMPRISING A GENE ENCODING A SUPEROXIDE DISMUTASE
JPH10508743A (en) 1994-09-09 1998-09-02 ニューロクライン バイオサイエンシーズ, インコーポレイテッド Interleukin-1 type 3 receptor
FR2724846B1 (en) 1994-09-27 1996-12-20 Rhone Poulenc Rorer Sa METHOD OF TREATING CANCER BY REGULATING THE ACTIVITY OF RAS PROTEINS
FR2725726B1 (en) * 1994-10-17 1997-01-03 Centre Nat Rech Scient VIRAL VECTORS AND USE IN GENE THERAPY
JP3565859B2 (en) 1994-10-28 2004-09-15 ザ・トラステイーズ・オブ・ザ・ユニバーシテイ・オブ・ペンシルベニア Improved adenovirus and uses 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
WO1996014837A1 (en) 1994-11-09 1996-05-23 Genetic Therapy, Inc. Gene therapy for hypercholesterolemia
ATE195555T1 (en) 1994-11-29 2000-09-15 Takara Shuzo Co METHOD FOR PRODUCING TRANSFORMED CELLS
FR2727867B1 (en) 1994-12-13 1997-01-31 Rhone Poulenc Rorer Sa GENE TRANSFER IN MEDULLAR MOTONURONES USING ADENOVIRAL VECTORS
ATE278794T1 (en) * 1995-06-15 2004-10-15 Crucell Holland Bv PACKAGING SYSTEMS FOR HUMAN, HUMAN ADENOVIRUSES, FOR USE IN GENE THERAPY
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
US5837511A (en) * 1995-10-02 1998-11-17 Cornell Research Foundation, Inc. Non-group C adenoviral vectors
JP2001520511A (en) * 1995-12-08 2001-10-30 ザ ユーナヴァーサティ オブ アラバマ アト バーミングハム リサーチ ファンデーション Targeting adenovirus vector
WO1997024453A1 (en) 1995-12-28 1997-07-10 Chiron Corporation Receptor specific chimeric viral surface polypeptides for viral and particle incorporation and internalization in target cells
WO1997038723A1 (en) 1996-04-16 1997-10-23 Immusol Incorporated Targeted viral vectors
WO1998011221A2 (en) 1996-09-13 1998-03-19 Dana-Farber Cancer Institute Car, a novel coxsackievirus and adenovirus receptor
CA2266342C (en) 1996-09-25 2010-06-08 Novartis Ag Packaging cell lines for use in facilitating the development of high-capacity adenoviral vectors
US5877011A (en) * 1996-11-20 1999-03-02 Genzyme Corporation Chimeric adenoviral vectors
ATE296117T1 (en) * 1997-03-07 2005-06-15 Wistar Inst USE OF ADENOVIRAL VECTORS EXPRESSING PDGF OR VEGF TO HEAL TISSUE DEFECTS AND TO INDUCE HYPERVASCULISM IN MAMMAL TISSUES
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
IL132673A0 (en) 1997-05-08 2001-03-19 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
US20030017138A1 (en) 1998-07-08 2003-01-23 Menzo Havenga Chimeric adenoviruses
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
IL133032A (en) 1998-11-20 2007-06-03 Introgene Bv Adenoviral gene delivery vectors provided with a tissue tropism for smooth muscle cells and /or endothelial cells
ATE403006T1 (en) 1999-03-04 2008-08-15 Crucell Holland Bv USE OF AN ADENOVIRUS VECTOR FOR TRANSDUCING SYNOVIAL CELLS
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
US6913922B1 (en) 1999-05-18 2005-07-05 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US20050232900A1 (en) 1999-05-18 2005-10-20 Crucell Holland B.V. Serotype of adenovirus and uses thereof
WO2001083797A2 (en) 2000-04-28 2001-11-08 Avigen, Inc. 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
CA2421864A1 (en) 2000-09-20 2002-03-28 Crucell Holland B.V. Transduction of dendritic cells using adenoviral vectors
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
PT1497438E (en) * 2002-04-25 2010-02-04 Crucell Holland Bv Means and methods for the production of adenovirus vectors
DK1497440T3 (en) * 2002-04-25 2008-12-01 Crucell Holland Bv Stable adenoviral vectors and methods of propagation thereof
AU2003288273A1 (en) 2002-10-23 2004-05-13 Crucell Holland B.V. New settings for recombinant adenoviral-based vaccines
NZ539813A (en) * 2002-12-17 2008-04-30 Crucell Holland Bv A replication defective recombinant adenovirus comprising a antigenic determinant of Plasmodium falciparum wherein the antigenic determinant is SEQ ID 6
DE602005015332D1 (en) * 2004-02-23 2009-08-20 Crucell Holland Bv Method for cleaning viruses
PL1802336T3 (en) 2004-10-14 2012-03-30 Crucell Holland Bv Malaria prime/boost vaccines
US20070010016A1 (en) 2005-03-11 2007-01-11 Mccelland Alan Gene transfer with adenoviruses having modified fiber proteins

Patent Citations (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US4517686A (en) * 1982-08-04 1985-05-21 La Jolla Cancer Research Foundation Polypeptide
US4578079A (en) * 1982-08-04 1986-03-25 La Jolla Cancer Research Foundation Tetrapeptide
US4589881A (en) * 1982-08-04 1986-05-20 La Jolla Cancer Research Foundation Polypeptide
US4792525A (en) * 1982-08-04 1988-12-20 La Jolla Cancer Research Foundation Tetrapeptide
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
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
US5585254A (en) * 1987-08-21 1996-12-17 University Of Colorado Foundation, Inc. Autonomous parvovirus gene delivery vehicles and expression vectors
US5403484A (en) * 1988-09-02 1995-04-04 Protein Engineering Corporation Viruses expressing chimeric binding proteins
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5571698A (en) * 1988-09-02 1996-11-05 Protein Engineering Corporation Directed evolution of novel binding proteins
US5204445A (en) * 1988-10-03 1993-04-20 The Scripps Research Institute Peptides and antibodies that inhibit integrin-ligand binding
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
US5861290A (en) * 1989-01-23 1999-01-19 Goldsmith; Mark A. Methods and polynucleotide constructs for treating host cells for infection or hyperproliferative disorders
US5223394A (en) * 1989-04-10 1993-06-29 Biogen, Inc. Recombinant dna molecule comprising lymphocyte function-associated antigen 3 phosphatidylinositol linkage signal sequence
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
US5474935A (en) * 1990-05-23 1995-12-12 The United States Of America As Represented By The Department Of Health And Human Services 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
US5521291A (en) * 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
US5547932A (en) * 1991-09-30 1996-08-20 Boehringer Ingelheim International Gmbh Composition for introducing nucleic acid complexes into higher eucaryotic cells
US5543328A (en) * 1993-08-13 1996-08-06 Genetic Therapy, Inc. Adenoviruses having modified fiber proteins
US5756086A (en) * 1993-08-13 1998-05-26 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
US5443953A (en) * 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
US5559099A (en) * 1994-09-08 1996-09-24 Genvec, Inc. Penton base protein and methods of using same
US5712136A (en) * 1994-09-08 1998-01-27 Genvec, Inc. Adenoviral-mediated cell targeting commanded by the adenovirus penton base protein
US5731190A (en) * 1994-09-08 1998-03-24 Genvec, Inc. Penton base protein and methods of using same
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
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
US6329190B1 (en) * 1995-11-28 2001-12-11 Genvec, Inc. Targetting adenovirus with use of constrained peptide motifs
US5922315A (en) * 1997-01-24 1999-07-13 Genetic Therapy, Inc. Adenoviruses having altered hexon proteins
US6878549B1 (en) * 1998-04-24 2005-04-12 Introgene B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
US6455314B1 (en) * 1998-09-11 2002-09-24 Genvec, Inc. Alternatively targeted adenovirus

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090110695A1 (en) * 2006-03-27 2009-04-30 Menzo Jans Emko Havenga Compositions Comprising a Recombinant Adenovirus and an Adjuvant
US11077156B2 (en) 2013-03-14 2021-08-03 Salk Institute For Biological Studies Oncolytic adenovirus compositions
US11130968B2 (en) 2016-02-23 2021-09-28 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
US11401529B2 (en) 2016-02-23 2022-08-02 Salk Institute For Biological Studies Exogenous gene expression in recombinant adenovirus for minimal impact on viral kinetics
US11813337B2 (en) 2016-12-12 2023-11-14 Salk Institute For Biological Studies Tumor-targeting synthetic adenoviruses and uses thereof

Also Published As

Publication number Publication date
US7968087B2 (en) 2011-06-28
US6929946B1 (en) 2005-08-16
US20090253207A1 (en) 2009-10-08

Similar Documents

Publication Publication Date Title
US7968087B2 (en) Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US7749493B2 (en) Chimeric adenoviruses
US6803234B2 (en) Gene delivery vectors with cell type specificity for primary human chondrocytes
EP1159438B1 (en) Use of adenoviral vector for synovial cell transduction
US20060104953A1 (en) Methods and means for enhancing skin transplantation using gene delivery vehicles having tropism for primary fibroblasts, as well as other uses thereof
AU2002315947A1 (en) Adenoviral vectors with cell type specificity for mesenchymal cells
EP1406666A2 (en) Adenoviral vectors with cell type specificity for mesenchymal cells
EP1020529B1 (en) Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
US7238528B2 (en) Means and methods for fibroblast-like or macrophage-like cell transduction
AU770780B2 (en) Gene delivery vectors provided with a tissue tropism for smooth muscle cells, and/or endothelial cells
EP1191105A1 (en) Gene delivery vectors provided with a tissue tropism for T-lymphocytes
EP1191104A1 (en) Gene delivery vehicles and use thereof in the preparation of a medicament and/or vaccine
NZ527634A (en) Means and methods for synoviocyte cell transduction
AU2001294348A1 (en) Transduction of chondrocytes using adenoviral vectors

Legal Events

Date Code Title Description
AS Assignment

Owner name: CRUCELL HOLLAND B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VOGELS, RONALD;HAVENGA, MENZO J.E.;BOUT, ABRAHAM;REEL/FRAME:018578/0448

Effective date: 20021127

STCB Information on status: application discontinuation

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