CA2198861A1 - Recombinant adenovirus comprising a chimeric penton base protein - Google Patents

Recombinant adenovirus comprising a chimeric penton base protein

Info

Publication number
CA2198861A1
CA2198861A1 CA002198861A CA2198861A CA2198861A1 CA 2198861 A1 CA2198861 A1 CA 2198861A1 CA 002198861 A CA002198861 A CA 002198861A CA 2198861 A CA2198861 A CA 2198861A CA 2198861 A1 CA2198861 A1 CA 2198861A1
Authority
CA
Canada
Prior art keywords
amino acid
acid sequence
penton base
gene sequence
chimeric
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
CA002198861A
Other languages
French (fr)
Inventor
Douglas E. Brough
Joseph T. Bruder
Imre Kovesdi
Duncan L. Mcvey
Thomas J. Wickham
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.)
Genvec Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2198861A1 publication Critical patent/CA2198861A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70546Integrin superfamily
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/14011Baculoviridae
    • C12N2710/14041Use of virus, viral particle or viral elements as a vector
    • C12N2710/14043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vectore
    • 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
    • C12N2710/14011Baculoviridae
    • C12N2710/14111Nucleopolyhedrovirus, e.g. autographa californica nucleopolyhedrovirus
    • C12N2710/14141Use of virus, viral particle or viral elements as a vector
    • C12N2710/14143Use 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/40Vectors comprising a peptide as targeting moiety, e.g. a synthetic peptide, from undefined source
    • 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
    • C12N2810/40Vectors comprising a peptide as targeting moiety, e.g. a synthetic peptide, from undefined source
    • C12N2810/405Vectors comprising RGD peptide
    • 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
    • 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
    • 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
    • C12N2810/80Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates
    • 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
    • C12N2810/80Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates
    • C12N2810/85Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian
    • C12N2810/855Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian from receptors; from cell surface antigens; from cell surface determinants
    • 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
    • C12N2810/80Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates
    • C12N2810/85Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian
    • C12N2810/855Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian from receptors; from cell surface antigens; from cell surface determinants
    • C12N2810/856Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian from receptors; from cell surface antigens; from cell surface determinants from integrins
    • 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
    • C12N2810/80Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates
    • C12N2810/85Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian
    • C12N2810/859Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian from immunoglobulins

Abstract

A recombinant adenovirus comprising a chimeric penton base protein, which includes a nonpenton base sequence, and a therapeutic gene, a method of gene therapy involving the use of such adenovirus, and adenoviral transfer vectors for the generation of such recombinant adenovirus are provided.

Description

w o 96/07734 ~ PcTrusg5/o99o4 RECOMBINANT ADENOVIRUS COMPRISING A CHIMERIC PENTON BASE PROTEIN

TECHNICAL FIELD OF THE INVENTION
The present invention relates to a recombinant adenovirus comprising a chimeric adenoviral penton base protein and the use of a recombinant adenovirus comprising a chimeric adenoviral penton base protein in gene therapy.
BACKGROUND OF TH~ INVENTION
Adenoviruses belong to the family Adenoviridae, which is divided into two genera, namely Mastadenovirus and Aviadenovirus. Adenoviruses are nonenveloped, regular icosahedrons 65-80 nm in diameter (Horne et al., J. Mol. Biol., 1, 84-86 (1959)). The capsid is composed of 252 capsomeres of which 240 are hexons and 12 are pentons (Ginsberg et al., Virology, 28, 782-783 (1966)).
The hexons and pentons are derived from three different viral polypeptides (Maizel et al., Virology, 36, 115-125 (1968); Weber et al., ViroloqY, 76, 709-724 (1977)). The hexon comprises three identical polypeptides of 967 amino acids each, namely polypeptide II (Roberts et al., Science, 232, 1148-1151 (1986)). The penton contains a z5 penton base, which is bound to tne capsid, and a flber, which is noncovalently bound to and projects from the penton base. The fiber protein comprises three identical polypeptides of 582 amino acids each, namely polypeptide IV. The Ad2 penton base protein is an 8 X 9 nm ring-shaped complex composed of five identical protein subunits of 571 amino acids each, namely polypeptide III
(Boudin et al., Virology, 92, 125-138 (1979)). Proteins IX, VI, and IIIa are also present in the adenovirus coat and are thought to stabilize the viral capsid (Stewart et al., Cell, 67, 145-154 (1991); Stewart et al., EMBO J., 12(7), 2589-2599 (1993)).
Once an adenovirus attaches to a cell, it undergoes receptor-mediated internalization into clathrin-coated W096/07734 ~ ~ ~ 8 8 ~ ~ PCT~S95/09904 endocytic vesicles of the cell (Svensson et al., J.
Virol., 51, 687-694 (1984); Chardonnet et al., Virology, 40, 462-477 (1970)). Virions entering the cell undergo a stepwise disassembly in which many of the viral structural proteins are shed (Greber et al., Cell, 75, 477-486 (1993)). During the uncoating process, the viral particles cause disruption of the cell endosome by a pH-dependent mechanism (Fitzgerald et al., Cell, 32, 607-617 (1983)), which is still poorly understood. The viral particles are then transported to the nuclear pore complex of the cell (Dales et al., Virolo~y, 56, 465-483 (1973)), where the viral genome enters the nucleus, thus initiating infection.
An adenovirus uses two separate cellular receptors, both of which must be present, to attach to and efficiently infect a cell (Wickham et al., Cell, 73, 309-319 (1993)). First, the fiber protein attaches the virus to a cell by binding to an, as yet, unidentified receptor. Then, the penton base binds to ~ integrins.
Integrins are a family of heterodimeric cell surface receptors that mediate cellular adhesion to the extracellular matrix molecules fibronectin, vitronectin, m; ni n, and collagen, ag well as other molecules (Hynes, Cell, 69, 11-25 (1992)). Integrins are known to play important roles in cell signaling processes, including calcium mobilization, protein phosphorylation, and cytoskeletal interactions (Hynes, supra). The specificity with which an integrin binds to a particular ligand, such as those associated with an adenovirus, is a function of the paired ~ and ~ subunits of the integrin.
For example, integrin ~6~1 binds to lAm- n~ n, integrin ~2~1 binds to collagen and lAm; n; n, and integrin ~3~1 binds to collagen, lam;n;n, and fibronectin. Furthermore, different tissue types may have different complements of integrin subunits, thereby providing a mode of spatial control over integrin-ligand signal transfer or over ligand internalization. Accordingly, some integrins, W096/07734 ~ ~ ~ 8 8~1 PCT~S95/09904 such as those that include subunit ~v~ are broadly expressed on numerous cell types, whereas other integrins have a much more narrow tissue distribution. For - example, the integrins that include subunit ~2 are expressed only on leukocytes, such as neutrophils and macrophages, integrins including subunit ~4 are expressed only on lymphocytes and fibroblasts, and the integrin defined by the subunits a~3 is expressed only on platelets and megakaryocytes.
The specificity of integrin subunit complement also extends to the infectability of cells by different serotypes of adenovirus, because the particular ~ and ~
subunits dictate whether a virus can enter a cell. For example, the penton base of the adenovirus serotype Ad2 binds to integrins ~v~3 and ~v~S (Wickham et al. (1993), supra). Given that both receptors utilized by an adenovirus are expressed on most human cells, nearly all cells in a human body are susceptible to adenoviral infection.
A majority of integrins have been found to recognize short linear stretches of amino acids in binding to a specific ligand. The tripeptide motif arg-gly-asp (RGD), which is found in scores of matrix ligands, including laminin, fibronectin, collagen, vitronectin, and fibrinogen, has been implicated in the binding of ~3~1 ~5~ IIb~3~ ~m~2/ and most, if not all, of the five ~v~
containing integrins. The conformation of the RGD
sequence within a matrix ligand is thought to be a primary factor in integrin specificity (Pierschbacher et al., J. Biol. Chem., 262, 17294-17298 (1987)). Sequences that directly flank the RGD sequence have been shown to influence integrin specificity, presumably because of their effect on RGD conformation (Smith et al., Proc.
Natl. Acad. Sci. USA, 90, 10003-10007 (1993)). However, sequences distant from the RGD may also function in integrin specificity as has been shown for the binding of ~5~1 to fibronectin (Obara et al., Cell, 53, 649-657 RECTiFlED SHEET (RU' E 91 ISAIEP

8 ~ ~
.

(199~). Other int~grlns, which do not utili~e RGD, have been ~ound to bin~ similar 9hort llnear ~tretches ~f a~i~o acids ~ithln t~eir specif c l~gands. For example.
i~tegrin ~3 bind~ via the amino acid 3equence ly3-gln-ala-~ly-asp (KQA~) [SEQ ID NO:l~ in ~ibrinogen ~Kloczewiak et al., Biochemi9trY, 23, 1767-1774 (19~4)), while ~l bind~ via the core s~q~ence glu-ile-leu-asp-val (~ILDV~ ~SEQ ID NO:2] in fibronectin (Komorlya ee al., ~.
Biol. ~hem., 266, 15075-15079 (l~gl) ) . A structural motif (i.e., asn-pro-xaa-tyr (NPXY) ) present in the subunits of ~V-containing i~tegrins has been ~hown to be impo~tant fo~ internalization tSuzuki et al., Proc. ~a.tl.
Acad. Sci. USA, 87, 5354 (1990)).
The penton base se~uence i~ highly conser~ed amons serotypes of adenoviru8 and contains five copies of the R~D tripe~tide ~otlf (~eu~ann e~ al., Gene, 6~, 15~-157 (19~ . The RGD tr peptide i~ belie~ed to mediate binding to a~ inte~rins because exosenously added RG~
peptides ~an block pentcn ~a~e ~inding and adenoviral
2~ infec~icn (Wickha~ et al. (1993), su~ra), and adenovlrus~3 that have point ~uta~ion~ in the RGD
~equence of the penton base are restricted in t~eir ability to infect cells (Bai et al., J.. ~i~ol., 67, 51~8-5205 ~19g3)).
The pen~on ba~e gene~ from Ad2, Ad5, Adl2~ and Ad40 serotyge~ of adenovirus have been sequenced. Alignment of the ~equences reveals a high degree of con~er~a~ion over the entire sequence, except for the N-terminu~, and, in Ad2, Ad~, and Adl2, a hyperva~iable region that
3 0 includes the RGD ~equence. Only Ad40, cne of two enteric adenoviral serotype6, doe~ not ha~e an RGD sequence. Ad2 and ~d5 are identical in t~e hypervariable region and contain a large insert of amino acids flanking either ~ide of the RGD sequence. Secondary structural analysis o~ the hypervarlable re~icn~ of the three RG~-c~ntainlng penton ~ases predicts that, in each ca~e, the RGD i9 flanked by ~-helice6. Such 3trUcture~ are belieYed to AMEN~ED S~IFET

W096/07734 pcT~s9slo99n4 form the spikes seen in cryo-electron micrographic (cryo-EM) images of Ad2 penton bases (Stewart et al. (1993), supra).
Recombinant adenoviral vectors have been used for the cell-targeted transfer of one or more recombinant genes to diseased cells or tissue in need of treatment.
Such vectors are characterized by the further advantage of not requiring host cell proliferation for expression of adenoviral proteins (Horwitz et al., In Viroloqy, Raven Press, New York, vol. 2, pp. 1679-1721 (1990); and Berkner, BioTechniques, 6, 616 (1988)), and, if the targeted tissue for somatic gene therapy is the lung, these vectors have the added advantage of being normally trophic for the respiratory epithelium (Straus, In Adenoviruses, Plenan Press, New York, pp. 451-496 (1984)).
Other advantages of adenoviruses as potential vectors for human gene therapy are as follows:
(i) recombination is rare; (ii) there are no known associations of human malignancies with adenoviral infections despite common human infection with adenoviruses; (iii) the adenoviral genome (which is a linear, double-stranded DNA) can be manipulated to accommodate foreign genes that range in size; (iv) an adenoviral vector does not insert its DNA into the chromosome of a cell, so its effect is impermanent and unlikely to interfere with the cell's normal function;
(v) the adenovirus can infect non-dividing or term1 n~ 1 ly differentiated cells, such as cells in the brain and lungs; and (vi) live adenovirus, having as an essential characteristic the ability to replicate, has been safely used as a human vaccine (Horwitz et al. (1990), supra;
Berkner et al. (1988), supra; Straus et al. (1984), supra; Chanock et al., JAMA, 195, 151 (1966); Haj-Ahmad et al., J. Virol., 57, 267 (1986); and Ballay et al., EMBO, 4, 3861 (1985)).

w096/07734 PCT~S95/09904 The problem of using a recombinant adenovirus in gene therapy i9 that all cells that express the aforementioned two receptors used by the adenovirus to attach and infect a cell will internalize the gene(s) being administered - not just the cells in need of therapeutic treatment. Likewise, certain cells, such as lymphocytes, which lack the ~v integrin adenoviral receptors, will be severely impaired in the uptake of an adenovirus and will not be easily ~m~n~hle to adenovirus-mediated gene delivery. Accordingly, limiting adenoviralentry to specific cells and/or e~p~n~;~g the repertoire of cells ~m~nAhle to adenovirus-mediated gene therapy would be a significant improvement over the current technology. Targeted adenoviral gene delivery should expand the cells ~men~hle to gene therapy, reduce the amount of adenoviral vector that is necessary to obtain gene expression in the targeted cells, as well as reduce side effects and complications associated with increasing doses of an adenovirus, such as inflammation and the transfection of normal, healthy cells.
Attempts have been made to target a virus to specific cells by sterically blocking adenoviral fiber protein with antibodies and chemically linking tissue-specific antibodies to the viral particle (Cotten et al., Proc. Natl. Acad. Sci. USA, ~9, 6094-6098 (1992)).
Although this approach has demonctrated the potential of targeted gene delivery, the complexity and reproducibility of this approach present major hurdles blocking its application in clinical trials. The difficulties thus far encountered in targeting the virus by these methods involve the method o~ synthesis required, which is to make major alterations in the viral particles following their purification. These alterations involve additional steps that covalently link large molecules, such as polylysine, receptor ligands and antibodies, to the virus. The targeted particle complexes are not homogeneous in structure and their W096/07734 ~ 8 ~ 1 PCT~S95/09904 ._ efficiency is sensitive to the relative ratios of viral particles, linking molecules, and targeting molecules used.
The present invention seeks to overcome the problem 5 of lack of cell and tissue specificity of recombinant adenoviral gene therapy. It is an object of the present invention to provide a receptor-specific, preferably cell receptor-specific/tissue receptor-specific, recombinant adenovirus. A further object of the present invention is to provide means for generating such a recombinant adenovirus at the level of gene expression, thereby enabling purification of recombinant adenoviral particle~
by conventional techniques. Another object of the present invention is to provide a method of gene therapy involving the use of such a homogeneous adenovirus, without the need for additional components or further modification. These and other objects and advantages of the present invention, as well as additional inventive features, will be apparent from the following detailed description.

~RIEF SUMMARY OF THE lNv~NllON
The present invention provides a recombinant adenovirus comprising a chimeric penton base protein that 2S selectively binds to a given receptor, preferably a cell-specific/tissue-specific receptor, an adenoviral transfer vector comprising a recombinant penton base gene for the generation of a chimeric penton base protein, and a method of using a receptor-specific recombinant adenovirus comprising a therapeutic gene in gene therapy.

~RIEF DESCRIPTION OF THE FIGURES
Figure 1 is a partial restriction map of an - adenoviral transfer vector (pAT).
Figure 2 is a partial restriction map of the vector pRcP~5 (~RGD).

w096/07734 ~ PCT~S95/09904 DETAILED DESCRIPTION OF THE INVENTION
The present invention provides, among other things, a recombinant adenovirus comprising a chimeric penton base protein. The chimeric penton base protein comprises a nonpenton base amino acid sequence, which is specific for binding to a receptor, a receptor-specific antibody domain or epitope, in addition to or in place of a wild-type penton base amino acid sequence, preferably an amino acid sequence that is specific for binding to a receptor, most preferably an RGD amino acid sequence.
By "nonpenton base amino acid sequence" is meant any amino acid sequence that is not found in a wild-type penton base. Preferably, the nonpenton base sequence is less than ten amino acids, more preferably less than five amino acids, and most preferably about three amino acids.
By "RGD amino acid sequence" is meant the RGD amino acid sequence and the RGD amino acid sequence along with up to or including three amino acids flanking either or both sides of the RGD amino acid sequence. The nonpenton base amino acid sequence renders the adenovirus specific for a given receptor, preferably a cell-specific or tissue-specific receptor, or specific for a given receptor-specific antibody domain or epitope. The receptor is preferably one that the wild-type adenovirus does not bind or one that the wild-type adenovirus binds but with greater specificity upon the introduction of the nonpenton base amino acid sequence into the chimeric penton base protein.
Preferably, the RGD amino acid sequence of the penton base protein has been replaced at the DNA level with an amino acid binding sequence for a given receptor, receptor-specific antibody ~omA;n or epitope.
Alternatively, the RGD amino acid sequence has been rendered inactive at the DNA level by mutation of the RGD
amino acid sequence, such as by insertional mutagenesis, for example, or rendered conformationally inaccessible in the penton base protein, such as by insertion of a DNA

W096/07734 ~ 8 ~ 1 PCT~S95/09904 sequence into or adjacent to the adenoviral penton base gene sequence, wherein "gene sequence" refers to the complete penton base gene sequence as well as any lesser gene sequences that are capable of being expressed as functional penton base protein. Preferably, the DNA
sequence is inserted near the gene sequence encoding the RGD amino acid sequence, 90 as to move the gene sequence encoding the RGD amino acid sequence within the penton base gene sequence such that in the ch;meric penton base protein the RGD amino acid sequence is conformationally inaccessible for binding to a receptor. In the latter case, the inserted nonpenton base gene sequence that causes the conformational inaccessibility of the RGD
amino acid sequence in the penton base protein is preferably one that encodes an amino acid sequence that is specific for binding to a receptor, a receptor-specific antibody ~m~l n or epitope. Such a recombinant adenovirus can be used, for example, to study receptor binding, adenoviral attachment, and adenoviral infection in vitro or in vivo.
In a preferred embodiment of the present invention, the above-described recombinant adenovirus additionally comprises a gene capable of being expressed in a cell to which the virus has attached or by which the virus has been internalized and preferably is one having therapeutic utility. In yet another preferred embodiment of the present invention, the recombinant adenovirus is fiberless or further comprises a chimeric coat protein, such as a fiber or hexon, that includes an amino acid sequence that is specific for a receptor, a receptor-~pecific antibody ~om~in or epitope, preferably one that is specific for the same receptor, antibody ~om~ln or epitope as the nonpenton base sequence. Such recombinant - adenovirus can be used, for example, to study the effects of expression of the gene in a given cell or tissue ln vitro or 1n vivo. Alternatively, the recombinant adenovirus can be used for gene therapy.

W096/07734 PCT~S95/09904 r The recombinant adenovirus comprising a chimeric penton base protein and the recombinant adenovirus that additionally comprises a gene-capable of being expressed in a particular cell can be generated by use of a viral transfer vector, preferably an adenoviral transfer (pAT) vector, in accordance with the present invention. The viral transfer vector, preferably the pAT vector, comprises a chimeric adenoviral penton base gene sequence. The chimeric penton base gene sequence comprises a nonpenton base sequence in place of the RGD
amino acid sequence, which has been deleted, or in addition to the RGD amino acid sequence, which has been mutated or rendered conformationally inaccessible in the expressed chimeric penton base protein as described above. The nonpenton base sequence renders the adenovirus specific for binding to a receptor, receptor-specific antibody domain or epitope also as described above. Given that the penton base gene has been shown to be ~90~ conserved among 5 of the approximately 41 serotypes of adenovirus, it is expected that any one of the serotypes of adenovirus may be used as the source of the penton base gene. It is preferred, however, that one of the serotypes for which the penton base gene has been sequenced is used.
Restriction sites that are unique with respect to the adenoviral genome are introduced into the penton base gene sequence; preferably, such restriction sites are introduced into or flanking the RGD region of the penton base gene sequence by a suitable method, such as PCR
mutagenesis. These unique restriction sites may be any restriction site not already present in the penton base gene, and are preferably Eco RI and/or Spe I. Such sites facilitate the removal, inactivation, such as by sequence alteration, of the DNA sequence encoding the RGD amino acid sequence in a given adenoviral genome, such as a wild-type adenovirus, or the rendering of the RGD amino acid sequence conformationally inaccessible, thereby W096/07734 ~ 6 ~ PCT~SsS/o99o4 ._ altering or eliminating the ability of the penton base molecule to bind an ~v integrin receptor. A deleted RGD
amino acid sequence can be replaced with, or a mutated or conformationally inaccessible RGD sequence can be accompanied by, a different DNA sequence, preferably a DNA sequence encoding specificity for binding to a receptor, preferably a cell-specific or tissue-specific cell-surface receptor, or to a receptor-specific antibody domain or epitope, for example.
Preferably, the pAT vector is one into which any suitable receptor-specific sequence can be rapidly inserted. For example, a unique Spe I restriction site can be used to remove the RGD amino acid sequence.
Alternatively, sequences also can be inserted into the penton base gene sequence without the need for unique restriction sites through PCR. Becau~e a recombinant adenovirus can be created via ligation of recombinant sequences with viral DNA or via homologous recombination, the pAT vector preferably has either (1) unique restriction sites that allow ligation of a vector fragment with the complementing fragments of the re~lning viral genomes, as described in Example 4, or (2) adequate lengths of DNA on either side of the receptor-specific or antibody domain- or epitope-specific sequence that allow efficient homologous recombination with viral DNA, as described in Example 5. A preferred pAT vector is shown in Figure 1, which is a partial restriction map of such a vector. The pAT of Figure 1 was generated as described in Example 1.
DNA encoding short peptide sequences or protein dom~nc capable of binding to a given receptor, preferably a specific cell or tissue receptor, and capable of being internalized by the receptor, such as the receptor that is resident on leukocytes, is preferred for insertion into the penton base gene sequence in which the RGD amino acid sequence has been deleted, mutated, or rendered conformationally inaccessible. However, other W096l07734 PCT~S95/09904 8 ~ 61 DNA sequences, such as those that encode receptor-specific antibody dom~n~ and sequences that encode antigenic epitopes recognized by specific antibodies also may be used to replace the RGD amino acid sequence.
The size of the DNA used to replace the RGD amino acid sequence may be constrained, for example, by impeded folding of the penton base and improper assembly of the penton base/fiber complex. However, cryo-EM analysis of the penton base indicates that, if the RGD amino acid sequence exists at the spikes of the penton bases, there may be considerable room to accommodate large inserts without folding or assembly being affected adversely.
Also, the large differences in size between the hypervariable region of the Ad5 penton base and the Adl2 penton base indicates that the hypervariable region may be able to accommodate inserts of various sizes. Of course, assembly of the penton complex probably will not be an issue with respect to viruses that are constructed without a fiber molecule.
The target receptor can be any receptor that is internalized by a cell, is optimally cell-specific or tissue-specific, and desirably is expressed only on those cells or tissue to be treated. Tissue-specific integrins are preferred target receptors for chimeric penton base molecules for four reasons. First, many integrins bind ligands via short linear stretches of amino acids, and these stretches have been identified for many integrins.
Second, certain integrins are tissue-specific for cells ~mPn~hle to targeted gene therapy. For example, ~2-cont~;n~ng integrins are expressed only in leukocytes,thus allowing targeted delivery to normally non-susceptible target cells while also preventing delivery to normally susceptible non-target cells. Third, the wild-type penton base uses ~v integrins to enter cells so that the use of a related integrin molecule is more apt to be successful than the use of an unrelated receptor.
Fourth, integrins are efficiently internalized into the W096/07734 ~ ~ ~ 8 ~ 6 1 PCT~S95/09904 cell (Bretscher, EMB0, 11, 405-410 (1992)), which is required for viral infection of cells.
Changing the penton base protein such that it recognizes a receptor, such as a cell-specific or tis~ue-specific cell-surface receptor, typically will not alter viral particle attachment to cells because cell attachment is mediated by the adenoviral fiber protein.
Infection of the cells to which the chlmeric penton base protein has attached, however, will be generally impeded because the virus requires the presence of an ~v integrin to enter the cell. Therefore, an adenovirus with altered penton bases will bind to most cells but will enter only those cells expressing the receptor (e.g., the integrin) for the chimeric penton base. Those cells unable to express the appropriate receptor nonetheless will have adenoviral particles tethered on the cell membrane because the viral particles will be unable to enter the cell. Consequently, tethered adenoviral particles will be exposed to the immune system and disposed of accordingly.
Following fiber-mediated viral attachment to cells, it is possible that some virus may manage to non-specifically enter cells not expressing the cell- or tissue-specific receptor recognized by the chimeric penton base. Therefore, some leakiness in cell-specific or tissue-specific expression may be observed. However, if this is the case, fiber-deficient viral particles can be constructed that use the penton base as the primary cell attachment protein. It is known that fiber protein is nonessential for virion assembly or infectivity of cells (Falgout et al., J. Viroloqy, 62, 622-625 (1988)).
Such mutant adenoviral particles lacking fiber protein adsorb onto cells at a ten-fold lower rate. The fact that the mutant viruses do adsorb and at a lower rate than wild-type particles indicates that the adsorption is mediated by the penton base/Q~ integrin interaction, which has a 30-fold lower affinity than the fiber/fiber-W096/0773~ 8 ~ ~ PCTNS9~5~1 receptor interaction (Wickham et al. (1993), ~upra).
Accordingly, both attachment and internalization can be restricted only to those cells expressing the desired tissue-specific receptor.
By replacing the low affinity RGD amino acid sequence in a wild-type penton base with an amino acid sequence with a high affinity for a given receptor, such as a cell-specific or tissue-specific receptor, it is possible to generate a fiberless viral particle with as high, or higher, affinity for targeted cells than the wild-type fiber-expressing virus. Fiberless viral particles containing a chimeric penton base, compared to fiber-cont~in'ng viral particles, can be targeted more effectively to tissues because the altered virus is able to bind only to the cells expressing the cell-specific or tissue-specific receptor recognized by the chimeric penton base. The desired ligand and receptor characteristics are as described above.
A recombinant ~hlmeric penton base gene sequence can be moved from a pAT vector into baculovirus or a suitable prokaryotic or eukaryotic expression vector for expression and evaluation of receptor specificity, avidity, and other biochemical characteristics.
Accordingly, the present invention also provides recombinant baculoviral and prokaryotic and eukaryotic expression vectors comprising a chlm~ric adenoviral penton base gene sequence. The chimeric penton base gene sequence includes a nonpenton base sequence in addition to or in place of a wild-type penton base amino acid sequence, such as an RGD amino acid sequence, which is specific for binding to a receptor. The wild-type penton base amino acid sequence may be deleted, mutated, or rendered conformationally inaccessible as described above with respect to the recombinant adenovirus comprising a chimeric penton base protein. By moving the chimeric gene from pAT to baculovirus or a prokaryotic or eukaryotic expression vector, high protein expression is W096/07734 ~ 8 PCT~S95/09904 . ~;'1 achievable, resulting in approximately 50~ of .he total protein of the host being the chimeric penton base.
Accordingly, the present invention also provides a recombinant baculovirus comprising a chimeric penton base protein and a chimeric adenoviral penton base protein comprising a nonpenton base amino acid sequence in addition to or in place of a wild-type penton base amino acid sequence, such as an RGD amino acid sequence, which is specific for binding to a receptor. The nonpenton base amino acid sequence is specific for binding to a receptor or a receptor-specific antibody domain or epitope as described above. For protein characterization studies, the recombinant chimeric penton base protein (rcPB protein, such as rcPB5) can be purified using any suitable methods, such as those described by Wickham et al. (1993), supra.
Various characteristic parameters of the penton base protein of interest can be assessed as follows: Adhesion assays are used to evaluate the specificity of the interaction of the rcPB protein with its designated receptor, using the method of Wickham et al. (1993), supra, for example. Specificity and affinity of the receptor/rcPB interaction is assessed by Scatchard analysis as shown previously by Wickham et al. (1993), supra, for wild-type penton base protein. Receptor specificity is further assessed by using antibodies and peptides specific for the targeted receptor to block rcPB5 binding to cells, using conventional methods.
Internalization is assayed with rcPB protein and recombinant adenoviruses, e.g., Ad5 rcPB, as described previously by Wickham et al. (1993), supra, for example.
- rcPB binding to fiber protein is assessed by its ability to precipitate radiolabeled fiber molecules when coupled - to protein A-coated beads via an antibody to the penton base molecule. Alternatively, fiber binding can be assessed in an ELISA-based assay in which rcPB is coated onto ELISA plates followed by incubation of immobilized W096/07734 ~ 6 ~ ~CT~S95/09904 rcPB with soluble fiber molecules, using conver,tional methods. Fiber binding to the rcPB protein is then assessed by a further incubation with antibody specific for the fiber molecule, via conventional techniques.
Virus entry and gene expression are evaluated initially by using the pAT vector cont~;n;ng the insert of interest to generate recombinant virus expressing the chimeric penton base protein and a marker gene, such as ~-glucuronidase. ~-glucuronidase expression in cells infected with adenovirus cont~;n;ng the ~-glucuronidase gene (Ad-Gluc) can be detected as early as two hours after adding Ad-Gluc to cells. This procedure provides a quick and efficient analysis of cell entry of the recombinant virus and gene expression, and is implemented readily by an artisan of ordinary skill using conventional techniques.
A recombinant virus, which lacks a wild-type receptor binding sequence, such as the RGD amino acid sequence, in the penton base protein, can be produced in human embryonic cell line 293 (HEK 293), which has been shown to be able to replicate recombinant adenoviral particles lacking the RGD sequence, using previously described techniques (Bai et al. (1993), supra). A
recombinant virus, which lacks functional fiber proteins and in which the penton base acts as both the viral attachment and internalization protein, may be produced in cell lines which express the receptor to which the rcPB5 protein is targeted. For example, Ad5 rcPB5 cont~;n;ng an insert specific for ~3 can be produced in HeLa cells that express ~3 or 293 cells transfected with the gene for ~B3 .
The chimeric penton base protein may be present in the viral particle with the fiber, without the fiber (in which case the rcPB protein functions as the virus attachment and internalization protein), with a recombinant fiber molecule that is specific for a receptor, such as a cell-specific or tissue-specific W096/07734 ~ PCT~S9S/09904 receptor, or with a recombinant coat protein that is specific for a receptor, such as a cell-specific or tissue-specific receptor. See Examples 4-9 for a further description of these embodiments of the invention.
Recombinant adenoviruses of the present invention can be used to treat any one of a number of diseases by delivering to targeted cells corrective DNA, i.e., DNA
encoding a function that i8 either absent or impaired, or a discrete killing agent, e.g., DNA encoding a cytotoxin that, for example, is active only intracellularly, or DNA
encoding ribozymes or antisense molecules, for example.
Accordingly, use of the term "therapeutic gene" is intended to encompass these and other embodiments of that which is more commonly referred to as gene therapy and is known to those of skill in the art. Diseases that are candidates for such treatment include, for example, cancer, e.g., melanoma or glioma, cystic fibrosis, genetic disorders, and pathogenic infections, including HIV infection. For example, a recomhinant adenovirus having a penton base molecule recognized by ~ 3 receptors can be used to treat melanoma or glioma, and a recom.binant adenovirus recognized by ~3~1 receptors and expressing the cystic fibrosis tr~ncm~mhrane regulator (CFTR) gene can be used to treat cystic fibrosis by delivery to the epithelial cells of the lungs.
Furthermore, various blood-related diseases can be treated by using a recombinant adenovirus recognized by Qm~2 receptors to target neutrophils and macrophages, a recombinant adenovirus recognized by ~4~1 receptors to target lymphocytes, a recombinant adenovirus recognized by ~IIb~3 receptors to target plateletc and - megakaryocytes, and a recombinant adenovirus recognized by ~3 integrins to target endothelial cells undergoing anglogenesls .
One skilled in the art will appreciate that suitable methods of ~m~ni stering a recombinant adenovirus of the present invention to an ~nlm~l for purposes of gene W096107~34 ~ PCT~S95/09904 therapy (see, for example, Rosenfeld et al., Science, 252, 431-434 (1991); Jaffe et al., Clin. Res., 39(2), 302A (1991~; Rosenfeld et al., Clin. Res., 39(2~, 311A
(1991); Berkner, BioTechniques, 6, 616-629 (1988)), chemotherapy, and vaccination are available, and, although more than one route can be used to ~m;n; ster such a recombinant adenovirus, a particular route can provide a more immediate and more effective reaction than another route. Pharmaceutically acceptable excipients are also well-known to those who are skilled in the art, and are readily available. The choice of excipient will be determined in part by the particular method used to ~mln; gter the recombinant adenovirus. Accordingly, there is a wide variety of suitable formulations for use in the context of the present invention. The following methods and excipients are merely exemplary and are in no way limiting.
Formulations suitable for oral ~m~n;stration can consist of (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as solids or granules; (c) suspensions in an appropriate liquid; and (d) suitable emulsions.
Tablet forms can include one or more of lactose, mannitol, corn starch, potato starch, microcrystalline cellulose, acacia, gelatin, colloidal -~ilicon dioxide, croscarmellose sodium, talc, magnesium stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, moistening agents, preservatives, flavoring agents, and ph~rm~cologically compatible excipients. Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the W096/07734 ~ PCT~S95/09904 like containing, in addition to the active ingredient, such excipients as are known in the art.
The recombinant adenovirus of the present invention, alone or in combination with other suitable components, can be made into aerosol formulations to be ~m; n; stered via inhalation. These aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.
They may also be formulated as pharmaceuticals for non-pressured preparations such as in a nebulizer or an atomizer.
Formulations suitable for parenteral ~m; n; stration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The formulations can be presented in unit-dose or multi-dose sealed cont~;ners~ such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
Additionally, the recombinant adenovirus of the present invention may be made into suppositories by mixing with a variety of bases such as emulsifying bases - or water-soluble bases.
Formulations suitable for vaginal ~m; n; stration may - be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas cont~;n;ng~ in addition to the active ingredient, such carriers as are known in the art to be appropriate.

W096/0773~ 2 ~ PCT~S551~3~1 with the gene of interest, the composition employed, the method of administration, and the particular site and organism being treated. However, the dose should be sufficient to effect a therapeutic response.
In addition to the recombinant adenovirus of the present invention, the adenoviraI transfer vector also has utility in vitro. It can be used as a research tool in the study of adenoviral attachment and infection of cells and in a method of assaying receptor-ligand interaction. Similarly, the recombinant penton base protein comprising a nonpenton base amino acid sequence in addition to or in place of a wild-type receptor binding sequence, preferably the RGD sequence, can be used in receptor-ligand assays and as adhesion proteins in vitro or in vivo, for example.
The following examples further illustrate the present invention and, of course, should not be construed as in any way limiting its scope.

Exam~le 1 This example describes the construction of the adenoviral transfer vector (pAT) for making chimeric penton base molecules.
pAT, a partial restriction map of which is shown in Figure 1, was created by cloning the unique Bam HI/Pme I
fragment (13259-21561) from the Ad5 genome into pNEB 193 Rl-, a minor derivative of pNEB 193 (New England Biolabs, Beverly, MA), from which the unique Eco ~I restriction site was removed (SEQ ID NO:5~. The resulting vector was called pNEB 193 Rl- [Ad5 (Bam HI/Pme I)].
Two pairs of PCR primers were synthesized and used to amplify a region upstream (left side) and a region downstream (right side) from the RGD sequence. A unique Spe I site was inserte~ in~ the antisense primer (A5a(15147)S, SEQ I~ NO:14) u~ed to amplify the upstream region between the R~ sequence and a Bst XI site (15017). The Bst XI site is 35 bp upstream from the RECTIFIED SHEET (RULE 91) ISA/EP

W096/07734 ~ 8 ~ ~ PCT~S95/09904 penton base start codon (15052). A unique Spe I site was alsQ ~serted into the sense primer (A5a(15204)S, SEQ ID
~0:13~ to amplify the right side of the gene between the ~G~ amino acid sequence and an Asc I site (15671). The antisense primer (A5a(15659)) used ta ~plify the right side of the gene is shown as SEQ I~ NO:1 The sense primer (A5s(14153)E; SEQ ID NO:11) for the left side of the gene includes the naturaily occurring Bst XI site (15017) and also contains a unique Eco RI site 13 bp upstream from the penton base start codon, which is not in the original sequence. Using these primers, the left and right sides of the penton base gene were amplified and then cloned into pNEB 193 R1' [Ad5 (Bam HI/Pme I)] to create the pAT vector.
The net results of this cloning were the generation of a unique Eco RI site just upstream of the penton base start codon (SEQ ID NO:3) to facilitate subcloning into a baculovirus transfer vector and to facilitate the generation of a recom~inant adenovirus using the pAT
vector; the deletion of an 8 amino acid region (SEQ ID
NO:9) within the wild-type penton base containing the ~v integrin binding domain; and the incorporation of a unique Spe I site encoding the amino acids threo~i~e and serine in place of the deleted region (S~Q ID NO:~ so that alternative DNA sequences encoding receptor-binding amino acid motifs or other sequences of amino acids can be inserted at that site.

ExamPle 2 This example describes the construction of the base vector pRcPB5 (~RGD) for all cloning used to make a recombinant baculovirus for the expression of a recombinant chimeric Ad5 penton base protein (rcPB5 protein) and for making rcPB5 genes for incorporation5 into pAT and other related vectors.
pRcPB5 (^RGD), a partial restriction map of which is shown in Figure 2, was created by cloning the Eco RI/Bql RECTIFIED SHEET (RULE 91) ISA/EP

~- ~ S . ~ J .... ~ ; L '. L .. ~ ........ L .~
~ F 2 ~ ~ 8 ~ ~ ~

II fragmen~ from pAT intc the baculovirus transfer vec~or pAcSG2 ~pharmin~en~ San Diego, CA). A new sequence encoding a receptor binding damain is ln~erted into the unique Spe I Slt~ within the pen~on ba3e gene in pRcP~5 (~RGD). The re~lting vector i3 the~ u3ed to generate recombina~ baculovi~us con~truc~i so that rcPB5 Frotein can be generated in large quantit~es and e~aluated~
Based on evaluation of the rec~m~inant protein, the Eco RI/B~ ragment frsm the pRcP~5 deri~a~i~re i6 then cloned into pAT tO make the recombinant adencvirls (Ad5 rcP~).

Exa~le ~
T~is example describe~ the clonlng of integrin se~uences into t~e S~e I 5 'te of the pRcPB5 (~RG~) vector of Example 2.
A sequeAce recog~ized by a specifi~ integri~
recepror ig claned into t~e penton base cassette as follows. Overlapping sense and antisense oligcnuclectide~ encoding a desired integrin-specir~c sequence are synthesized. For example, the cverlapping sen6e ~Be~a 3RGD6, S~Q ID NO:l71 and antisense ~3eta 3RGDa, SEQ I~ ~O:l~) oligonucleotides that encode sequences that are speciic for the in~e~ri~ ~3 (SEQ ID
2~ NO:;5) a~d the overlap~ing ~en~e (~S-ls, SEQ ID NO:21) and antise~se (CS-la, S~Q ID NO:22) oligonucleotide that encode sequence~ that are specific for the integrin ~4~1 are sy~tne~ized. The ~en~e and antisense se~uence3 for each are mixed a~d co~verted to full-length double ~tranded ~equences ~y ~illing in the unpaired nucleotida~
with Klenow fragment. The resulting dau~le-stranded aequence ia then inserted into the Spe I site of the pRcPB5 (~RGD) ~ector following digestion with ~he endonuclea~es Xba I and SDe I. The resulting vec~ors, pRcP~5~a~3) and pRcpss ~4B1)~ are used t~ create t~e recombinant baculovirus ~ectors, AcNPV rcPB5~a~3~ and AcNP~ rcP~5(a4~l). The rcPB5 protein produced i~ in~ec~

At~EN~ED SHEET

W096/07734 ~ 8 ~ 1 PCT~S95/09904 recombinant baculovirus vectors, AcNPV rcPB5 (~3) and AcNPV rcPB5 (~4~1) . The rcPB5 protein produced in insect cells using the aforementioned baculovirus vectors is evaluated as described above. Based on the analyses in the baculovirus expression system, the Eco RI/Bql II
fragment from each of the vectors then is cloned into a pAT vector to produce the adenoviral vectors, pAT rcPB5 (~3) and pAT rcPBS (~4~1) .

Example 4 This example illustrates the creation of recombinant adenovirus particles containing a wild-type fiber protein and a chimeric penton base specific for ~4 integrins (Ad5 rcPB5 (~4) :wtFiber).
A chimeric penton base specific for ~4 integrin (Ad5 rcPB5 (~4) :wtFiber) can be constructed through ligation of plasmid DNA with viral DNA, which chimeric construct then can be transfected into cells using st~n~rd methods. The pAT vector cont~;n;ng the desired chimeric form of penton base is digested with Xmn I to generate a 1 kb piece of DNA cont~;n;ng the receptor-specific coding sequence. Ad5 DNA is digested with the restriction endonuclease Xmn I, which cuts wild-type Ad5 at positions 14561 and 15710 within the Ad5 genome. The two larger fragments are purified away from the smaller 1 kb piece cont~;n'ng the sequence encoding RGD and then ligated with the Xmn I fragment from the pAT vector. The ligated fragments are then transfected into the appropriate cell line to produce recombinant virus. The virus produced from the cells can be plaque purified and ~erified to contain the chtm~ric penton base gene u~ing PCR. The penton base produced in the Ad5-infected cells can be also purified, and its receptor-specificity verified.

Example 5 This example illustrates the creation of recombinant adenovirus particles containing wild-type fiber protein W096/07734 ~ 8 ~ ~ PCT~S9510~30 and chimeric penton base specific for ~4 integrins (Ad5 rcPB5 (~4) :wtFiber).
Adenovirus particles having a chimeric penton base specific for ~4 integrins (Ad5 rcPB5 (~4) :wtFiber) can be constructed through transfection of plasmid DNA and viral DNA into cells and generation of recombinants through homologous recombination. The pAT vector cont~;n~ng the desired chimeric form of penton base is linearized by digestion with Bam HI. AdS DNA is digested with the restriction endonuclease Xmn I which cuts wild type Ad5 at positions 14561 and 15710 within the Ad5 genome.
These two unique sites within the Ad5 genome flank the S~e I site in the pAT vector. The linearized pAT vector and the Xmn I-digested Ad5 DNA are then transfected into the appropriate cell line, such as 293 cells, to produce a recombinant virus by homologous recombination. The virus produced from such cells can be plaque purified and verified to contain the ch'meric penton base gene using PCR. The penton base produced in Ad5-infected cells also can be purified, and its receptor-specificity verified.

Example 6 This example illustrates the creation of recombinant adenovirus particles lacking fiber protein and chimeric penton base specific for ~4 integrins (Ad5 rcPB5 (~4 ):
-Fiber).
The Ad5 rcPB5 (~4) :wtFiber viral DNA can be digested with Bam HI and the 0-59 map unit (m.u.) fragment purified. The fiber gene can be deleted from plasmid cont~;nlng the Ad5 Bam HI/Sal I fragment (genome map units 59-100). The resultant plasmid is then digested with Bam HI and Sal I, ligated to the 0-59 m.u. piece of Ad5 rcPB5 (~4) :wtFiber DNA, and transfected into cells expressing ~4 integrins. The ~4 integrins can serve as both the attachment and internalization receptors for the Ad5 rcPB5 (~4) :-Fiber virus.

Example 7 This example iIl~3trate~ the creaticn of recombin~nt adeno~ u~ particles lack~ng fiber protein and containi~g chimeric penton base with an epitope reccgnized ~y a mcnocl~nal antibody ~a~tibodY-SPeC~iC sequence).
~ ira~ DNA is i301ated fro~ an Ad5 viru~ lacking fi~er, a3 in Example 6, and dige~t2d with ~he endonuclease X~n r as ~n ~xa~ple 5. The pAT ~ec~or containing tne chimeric penton base gene contai~ing t~e lo sequence encoding the anti~ody-speci~ic epitope i9 prepared a~ in Example 5. The dige~ted viral and pAT DNA
i~ transfected intc 253 cell~ to produce ~irus ky homolosous recomblnati~n. ~e recombinant ~ir~5 iS
passaged and a~plified by incubatin~ cell ly~aees with a ~ispecific an~ibody to av in~egrin~ ~expressed and ~nternalized in 2~3 cells) and the antibody-~pecific epitope ~n the chlmeric penton base, thereby allowing the recom~inant virus to attach and infect the 293 cells.
The virus proge~y from infected cell~ lack the bi~pe~ific antib~dy 50 th~t ~allowing their puriflcation fro~
infecte~ celL~ they can be targeted to ~IV-infected cells by combinin~ them wich a bispeci~ic antibody recognizing the antibody-specific epitope in the chimeric penton base and the HI~ gpl20.

Example ~
Thi~ example illustrates the creation of recombinant adenovirus particle~ containing chimeric fiber protein ~pecif ic for sialyl hewis X an~igen and cbimeric penton base s~ecific for ~4 integrins ~Ad5 rcPB5 (a4):rcFi~er The pr~tein P-selectin binds to ~iaIyl Lewis X
antigen present on lymphacyte~. The bi n~i n~ domain from P-sele~tin i9 pre~en~ on the rcFiber (~LX) protein to mediate viru3 binding. The left ar~ (0-59 m.u.) of the Ad5 r~PB5 (~4~:wtFiber YirU~ can be prepared ~y 3a~ HI
digestion and then purified.

A~IE~D SHE~

W096/07734 ~ 6 1 PCT~S95/09904 The rcFiber (sLX) gene can be cloned into the plasmid containing the 59-100 m.u. fragment of the Ad5 genome. The rcFiber (sLX) gene then can replace the wtFiber gene. The 59-100 m.u. Ad5 fragment containing rcFiber (sLX) then can be ligated to the 0-59 m.u. Ad5 fragment containing rcPB5 (~4). The ligated fragment then can be transfected into cells expressing the receptors for attachment (sialyl Lewis X) and internalization (~4 integrins) for the chimeric virus Ad5 rcPB5 (~4) :rcFiber (sLX).

Example 9 This example illustrates the creation of recombinant adenovirus particles containing chimeric hexon protein specific for sialyl Lewis X antigen, chimeric penton base specific for ~4 integrins, and lacking fiber protein [Ad5 rcPB5 (~4): -Fiber:rcHexon (sLX)].
The protein P-selectin binds to sialyl Lewis X
antigen present on lymphocytes. The binding domain from P-selection is present within an exposed loop on wtHexon, and the resultant rcHexon (sLX) protein can mediate virus binding. Genes encoding rcPB5 (~4) and rcHexon (sLX) can be cloned into the pAT vector that contains the penton base gene and the region encoding the exposed loop of hexon protein. A ligation of three fragments can be performed to generate the recombinant virus. Ad5 rcPB5 (~4) :-Fiber virus may be digested with Eco RI and Bam HI.
The left end of the genome up to the Eco RI site and the right end of the genome down to the Bam HI site can be purified and ligated with the Eco RI/Bam HI fragment from the pAT vector cont~1n~ng rcpBs (~4) and rcHexon (sLX).
The ligated fragment then can be transfected into cells expressing the receptors for attachment (sialyl Lewis X) and internalization (~4 integrins) for the chimeric virus Ad5 rcPB5 (~4) :rcFiber (sLX).

W096/07734 ~ PCT~S~5/~5v1 Example 10 This example describes the expression of wild-type Ad5 and mutant rcPB5 (~RGD) penton base protein in baculovirus.
s Using PCR, the wild-type Ad5 penton base sequence was amplified with an Eco RI site inserted at the 5' end of the gene. This gene was then cloned into the pAT
vector and then subcloned into the pAcSG2 baculoviral transfer vector, using methods known in the art.
Recombinant baculovirus encoding the Ad5 penton base gene (AcNPV wtP35) produced levels of penton base protein in insect cells that constituted approximately 50~ of the total cellular protein. The purified wtPB5 protein migrated consistently with respect to its known molecular weight on SDS-PAGE and can mediate cell adhesion like Ad2 penton base (Wickham et al. (1993), supra).
The core baculoviral transfer vector, pRcPB5 (^RDG), for expression of rcPB5 protein in the baculoviral expression system, has been used to make recombinant baculovirus, which expres~es rcPB5 protein. This rcPB5 protein, rcPB5 (ARGD), contains an 8 amino acid deletion in the RGD region as shown in Figure 2, which is a partial restriction map of the vector pRcPB5 (~RGD).
This protein migrates consistently with respect to its known molecular weight on SDS-PAGE and, in contrast to the wild-type protein (wtPB5), does not mediate cell adhesion, which i8 consistent with its lack of the RGD
cell adhesion motif. Chtmpric recombinant penton base molecules rcPB5 (~3) and rcPB5 (~4), each cont~;ntng inserts ~pecific for ~V~3 and ~4 integrins, respectively, also have been produced via the core baculoviral transfer vector, pRcPB5 (~RGD). The rcPB5 (~3) protein mediates epithelial cell (~+, ~4- ), but not lymphocyte (ov-, ~4+), adhesion; in contrast, RcPB5 (~4) supports lymphocyte, but not epithelial cell, adhesion.

W096/07734 PCT~S95/09904 Example 11 This example illustrates the targeting of adenovirus to lymphocytes.
The transfer vectors pAT (~V~3) and pAT (~4) were created by cloning the Eco RI/Bgl II fragments from their respective pRcPB5 vectors into the pAT (~RGD) vector.
These pAT vectors were then linearized by Bam HI
digestion and transfected with Xmn I-digested viral DNA
from an Ad5 variant, which lacks the E1 and E3 regions and contains a ~-galactosidase reporter gene under control of a CMV promoter, to obtain recombinant Ad5 virus, namely Ad5 rcPB (~RGD), Ad5 rcPB ( ~ 3), and Ad5 rcPB (~4 ), through homologous recombination. The Xmn I
endonuclease cuts the wild-type Ad5 at positions 14561 and 15710 within the genome, while the Bam HI/Pme I
fragment contained in the pAT vector spans the Ad5 region of 13258 to 21562. Thus, the regions of homologous overlap between the left and right viral sequences with the Bam HI/Pme I fragment are 1303 bp and 5851 bp, respectively. The presence of the chimeric inserts in the Ad5 rcPB (~3) and Ad5 rcPB (~4) viruses are verified through PCR.
The recombinant adenovirus Ad5 rcPB (~4 ) is analyzed for its targeting potential in relation to wild-type Ad5 and recombinant Ad5 rcPB (~RGD) by measuring levels of the reporter gene expression in cells exposed to each of the viruses. Delivery of ~-galactosidase to the B-lymphocyte cell line, Ramos, is significantly higher using the Ad5 rcPB (~4) virus than the Ad5 rcPB (^RGD) and wild-type Ad5 viruses. Conversely, delivery of ~-galacto-qida~e to an epithelial-derived cell line i9 significantly higher than the wild-type Ad5 virus in comparison to the Ad5 rcPB (~4) and Ad5 rcPB (~RGD) viruses. These results ~mo~trate that changes in penton base protein that redirect the protein to interact with a new receptor can be utilized to target the virus w096/07734 ~ B ~ PCT~Sss/o99o4 _ to new cells and tissues while specifically avoiding delivery to other cells and tissues.

All publications cited herein are hereby incorporated by reference to the same extent as if each publication were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
While this invention has been described with emphasis upon preferred embodiments, it will be obvious to those of ordinary skill in the art that the preferred embodiments may be varied. It is intended that the invention may be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications encompassed within the spirit and scope of the appended claims.

SEQU~NCE ~9~ING

~; ~ Ga;~1ER~L INFo~MATIoN
( i) A~PLICANT: GEt~EC, In~-TrT~E OF ~ ;h,ioN: P~NTON 3RgE PROT}3IN AND hETHO~S OF ~SItlG SA~E
NUMBER OF SEQtlENCES: ~2 iv) COMPU~ E~EADA~ls FO~M:
~A) ~SEDIU~ TYP!3: Floppy diaX
(B) C:OMPU~ER: IH~ ~C co~pa~ible (C) OPE~lATINt~ SYST~ Pc-Dosll~s-l:)cs ~D~ SO~ E: PatentIn Releaae 1.0, Verolcn 1.25 ~PO~
v~ CD~h. APP~ICATION DATA:
tA) APPE~QTroN NUHB~: Wo pcTtus95/ossa4 ~8~ FI~IYG DATE: 01-AUG-L99, vi) PRIOR P.PPL~ lON DATA:
~A) APPLIQTI~N NU~ER: US 3a3162 ( 8 ~ FI ING DATE: OJ-S~3P-1994 2 ) INl~ 5ATION FOR SE~ ID NO: 1.
i~ 9E52UENC~5 CaMACTEP~ISTICS:
A ) ~NG1~1: 5 a~in~ acids (~ TYPE: amino acld (C) ST~ANDEDN~SS: ~ gle D ~ TOPOI.OGY: L lneaL~
~r ~C~ PrOt-in (Xi) 9EQU15NC~5 D3Sr~TPSION S~Q ID NO~1 r Yg G1n A1a G1Y A-P

( 2 ) INFORI~ATION FOR S~Q ID NO: 2:
U~ C~UAC~ STICS 1 ~A~ LI5NGTR: 5 am1nO aC~d~
(3) SYP~ nO aC1t ( C ) STR~U~UNe~g ~ ~ing1 ~ D ) TOP~ ne~r MOLE;C~ ; P~Otein ) SEQUBNC~ DBSCR~PT10-~: S~ rD Na:2:
G1U I1Q Leu A~p Val F~!r~fn ~HF~T ~

^ ~ t . ~; I .1. Li L . _ I J . `_,, " .~ . L ~

( Z ) INFOR~ TION EOR SEQ ~D NO: 3:
i ) Sl!~Qt~ÇNC2~ C~ CTERISTICS:
~1 LENGT~: ~S ba30 palrs ~B~ TYP~: nucl~ic acid C) S'rRANo~uN~SS ~ double ~D) T~P~r.OqY: llnlrn.~,,n ii ) MODECU~E TYPE; DNA ~ g~

~x) FE~AT~
A ) NAM~ S
~ B ) LOCATI:ON: 51. . 65 (xi) Si5~2~NC15 DBSC:RIPT~ON: SEQ ID NO: 3:
CTCACCl~AGG C:CATGGC~CC GAGC~ AT LCCCC~T ATG C~ 56 !Set ~q ~rg Aia Ala ( 2 ) :~NFQEWA~ oN FOR SBQ ID ~C: 4:
L) SEQIJ15NC~S CaM~CTER~SSrCS:
(A) r.ENGTH; 5 amino acid~
~B) TYPB~ a ~cit D ) ~OPODOGY: 1 Lnear ~ii) M~-T~'CUT~ TYP~: protei~
(xi) S~:QUENCD DESCRIPTrON; SEQ ID NO:~:
Me~ Arg Ala Ala S

12) l~ru -l~ssoN FO~I SISQ ID NO:5:
~i~ s~:o~ cE C~ARACSlS~rS~ICS:
~A) T3NGSE~ 65 ba~a pairs ( 9 ) ~PE: ~u~lel~ acid C ) Sl~ANDED~:SS: dou~l~
(D ) TOPOLOGY ~ r ( i~ ) ~r ~C~r ~ DNA ~ genomic ) ( iX ) Fl;ATURE
~A) r~3/ltEy; CDS
9 ) OCASSCN I 51. . 65 D S~F~T ~

. ~

(xi1 SEQU~NC~ DEsc~I2TToN: SEQ ~D NO:5:
CSCACCAA~G OcA~GGCAC~ GAGC~-T~ AT L'~CC~LAGT ATG C:~ 56 ~et ~g Arg ALa Ala ~2~ ~NFOR~ATION ~R SEQ ID NO:6:
( 1) SEQUENCB cHlu-a~ RIsTIcs: --tA~ L~al~: ~ a~ino acidc 3 ) TY?3~; amino acid D ) ~O~O~O&Y: 1 inear ( ii) ~OL~:C~ ~PE: protoin ( xl ) ~ ~ DESC~lPTION: SEQ ~D NO: 6:
g ala Ala 2 ~ INFORMATIO~ YOR SE~Q I3 NO2 7:
;uu~ s C~R~CT~UT~C~:
~A) 3ENGT~: 30 ba~e pair~
B ) T~ s nucleic acid ~C~ 5~ KCS: double (D ~ TO~WGY; Lnl~ .A
O~CU~S TYP25: DNA ~g-no~

( iX ~ FlSATU~:
~A~ Nl~/X~Y: CDS
B ) ~ 1..30 ~Xi I gEQlJUlCa~ D~3SC~ ON: Sl~ ~D ~10: 7:
GAC A~G AAC GAS AC~r AGT GCC ACa CGG G~ 30 A~p ~e~ A~n A~y ~hr ser Ala ~hr Arq Al~

( 2 ) INFO~TION POR ~lsq ~ NO~
( i ) S t~" b ~ CHA~Cl~RI8'TI CS:
~A) L3NGT~: 10 ~lno ~cidc ~B) SY~B: amLno acLd ~D) TOPOrOGy; llnear 1~3 M~OUT~ TYPB: protein : AMENl!ED SHEET

(xi ) S~;QU3NC~ DESC~IPSION: S~Q ID NO: ~:
Asp Het A~n Aep Thr Ser Ala Thr ~rg Ala NFOR!~ATION FOR SEQ ID NO:9:
( i ) SEQUISNCE ~MACTE~IsrIcs:
t~ LENG~H: ~ ba~e pairs ~B) TYP~3: nucl~i~ acid ( C ) STPAND~DNESS; do~bl~
D ) TOPOr OGY: unlc~awn Li ~ ~OLECU~E TYP~:: DNA ~ genomic ) ix ) FEA'rU~E;
( A ) NAY.E ¦ J~3Y: CD S
(8) ~ATION: 1..48 (xi) SEgu~5N-,c DE8C~IPTION: SEQ ID NO~
GAC ATG AAC GAT C~ GCC A~T CGC GGC GAC AC~ ~T GCC ACa CGG GCT 43 A~p ~et Aan Aap ~iA ~la Ile A~g Gly A~p 'Shr Phe Ala Thr Arg ~la ( 2 ) Lc~rO~ATION FOR SEQ ID NO: la ~L) ~ u~r~ ~p~ TsT~cs!
~A) r-ENOT~: 16 a~Lno acLdo ~) TYP13: amino acid ( D ) TopoI~oGr ~
r ~r ~ ~YPE: ~rotein ~xi~ SBQm~CB D~C~IPSION: SEQ I~ ~O: lO:
Acp Met Aan A~p ~i- Ala rI~ A~g Gly ~}j~ rhr ~ho Ala Shr Arg Ala 2 ) LN~ Io~ SISQ lD NC~
( ~ ~ g~U~ C;~ D~ I8TrCSl ~A) ~EIiG~EIs 5a ba~- p~irY
~S) TYPE: nuc1et c a~ld ( C ) ss~ANDEI~Rr:s9: ~inglc 0Pot~oGy ~ un Irn~ ,a nE~G~ TYPE: ~A (g~rJa~ic ) (xt ) SEQUENCB DESC5~IPTIOI~: SEQ ID NO:ll:
AM~CTQCC l~ CCr~TGG r~CCA~CG~ TG~.~ ,.. GAA.. 'C~CC;. TA 52 -t~ ~ st~

._ (2) INF~1~ASIC~N FOR SEQ ID NO:12:
v~ CT~SR~STICS:
( A ) ~ENGTH: 2 5 ba~e pai~
~) TYPE~ nucl~ic acid ~C) STE~RDEDN~35S: ~ ngle ~ ~ ) TOPO~O~;Y: unk~ wn IOL}~CU~E rYPE; DNA ~ gencmic ) ~xl~ SEQUENOE D~srgrpTIoN S~Q I~ ~:0:12:
CTGGa;GGCG CGCCA~AATC T~GTT 2 5 ( 2 ) I~F~ ATIor~ Fa~ ~Q ~D NO: 13:
t i ) S~4u ~ ctCE C~ARA~13R~STIC9 NGT~; 26 ba3~ paLr3 ~B) TYPE: nucleic acld ~ C ) S'rRANDlQ~NESS: 9ingle ( D ) TOPoLoGr: unkno~n (Li) I~ TYP2:~ DNA ~g~nomic) txL) S~QUl3~CF DESCRlPTION: S~Q ~D NO: 13;
~AAACrAGTG C~'~CACGGGC 'rGAG&A 2 ( 2 ) IN~IAT~ON JOR SSQ ID No: 14:
~i) SEQUENC~t CEARACTi~ISTICS:
~A) ~NG'r~: 26 ba~ pair~
~EI) TYPIC: nucleic acid C ~ S~AlilDleDNE~: ~ingle roPol,ocY ~ u~
SOE~CU~ SYP~: DNA (~r.:. ic~
(xL~ SEQUENCZ D~S~aTPSrQ~: S~Q ID No: 14;
A~AAI-T~-T~ ~ SGT CCSCCA 2 6 Z ) IN~A~ON PO~ S15~. ID NO:15:
i) SEQUENC$ C~ S~
~A~ L13NG~H: 68 ba~ palro ) TYP~: nuc lelc acid c ) STRA?~D~D~BSS: dol~b 1 c D ~ SOPOWGY un~cnown 11 ) MQ~,~:C~L~ ~YP~: DNA ~ geno~ic ) ix ) P~SATUR~ s ~A) ~a~lR~: CDS
t D ) ~OCATlON: 3 . . 68 bt~ ~, xi~ S~Q~ O~scR-pT7-oN: SEQ ID NO:15:
~A AC'r AGT G~;A GG~ ~GC TC~ TTT GGC CGC ~;GC GAC ASS CGC AAC ~GC 4 Thr ~er Gly Gly CyY Ser P!~e Gly Ar~ GLy A3p ;le Ar~ A~n Cy3 li GGC GGC CTG CAG TC'r AGA AAA 68 Gly Gly ~eu Gln Ser Arg Lye 2 ) INFOR~tATION E'C~ SEQ ID NO . 16:
(i) S~u~n~l: C~CTE:}~ISTrCS:
(A~ ~ENG~H: 22 a~ino acLda ~ 8 ) TYP~: a~lno acid ( D ~ 'rOPOI.06Y ~ Rar OI.i3CU~ TYPE: prot~i~
(xi~ s~Q-J. .~; D~SSCR~PTION: SEQ ~D Na.16:
rhr Ser Gly Gly Cya Ser Phe ~ly Arg Gly Asp Ile ~rg A~n Cy~ Gly Gly ~eu Gln S~r A~g ~yY

2 ) INFOR~ATrON FOR SEQ rD N<~- 17:
L) S2QU~3NC~3 CaARAC~IS'rrCS;
~A~ L~ENGT~ 41 ~a~-~ pairJ
3 ~ 5YPE: nucl~lc acid ~C~ S'lRANDXDNlSSS: ~ingle ~D ) TOPODXY ~ C..
~ Li ~ MOL~CU~ TY~B: D~A ( g~n 1 ~ ) (xL) SBQUINC~ D2~ IpTIoll: S~5Q ID NOtl7:
~AACTAGTGG AMASGCSCS ~ _ ~;CC~ GCGA~TSCG C 11 (2) IN~_LASIO~I FOR S~3Q ID ~la~
) S3QU~ c~cTERlsT~c3:
~A) L2NG~H: 42 bas~ pai.r~
nucl~lc a~d C ) STP~ ntn7-~s: ~i~gle t D ~ TOPOSO~Y ~
( i i ) ~SOL~:C~; TYPE: DNA ~ ~7~ - - i C ) (xi) Sl:QUENC9 DEgCRIPT~o~: ~BQ ID ~rc: 18:
AGAC ~;CA~;C~ CGCAGT~O~ AA~GTCGCCG CG 42 p,~JlE~3 S~

% ~ ~ ~

2 ) IN FORHATION FOR S~Q ID NO: 19:
u~ C~RACTERIST~cS;
~A) LE~GT~l: 110 ba~e pairs TYP~S: nucl~lc ~cid C ) STRA~ UP~ CS: doubl~
~1:1 ) Top~L~Gy 1~ nl~
OLLCl~ TYPE: DNA (~enc~m~c ) ?12ATUR~:
~A) ~ 13/XISY: CDS
t B ) LO~ATIQN: 3 . . 110 s3QuE~rcE DE;SC~IP~ION: SEQ ~D NO: l9:
~A AC~ AG'r ~;A GGA GAC GhG C~C ccA ~AG crc GTG ACS cTC C~ C 47 Thr s~ Gly Gly A~p Glu r~u Pro Gln ~e~ ~al Thr Le~ Pro Kl3 CQ AAC C~C CAC G~:T CCA GAG ATT CTC t:AC GSG CCA TCT ACT GGC GGC 95 Pro A~rl Leu Hi~ Gly Pro Glu Ile ~eu A~p val P~o .S~r Shr Gly GLy L~u Gln Ser Arg r.y~

l 2 ) T~ 2fATlON FOR SLQ LD NO: 20:
SEQt~NC:~ C~ rSTICS:
~A) ~ 36 a~ino aaid4 ~) TY~: a~lno ac~d ( D ) SO~
ii ) HO~ ~CU~ protein ~xi~ S~ 5NCL ~ CTP~ION; S~2 I~ NO:20:
hr Ser Gly GLy a~p G~u L--u Pr~ Gln ~eu Val Thr Leu ~r~ Eli~ ~r~
~n te~ Hin Gly Pro Clu Ile Leu A~p Val Pro 9cr Thr Gly Gly I,eu ln Ser Arg Ly~

~2~ F:lA'rION FOR 8E9 ID NO:21: _ L 1 S~QtJr',~ U~Y~T~c~:
~A) L~GTH: 65 ba~- palr~
(B) ~ I: nuc1~lc aci~
~ C ~ ST~ND~DN~
~o ) ToPo~oGr, ~ ~Q
~il) ~OI,~SCUT~15 ~ DNA ~g~ MFNnEn SH~T

L L . _ ~ J ~ lr~ J

(xi~ S~ n~ 0~5SC~IPT~O~: S~Q ID NO:21:
A~AcTAGrGG A'~ P^~CG~G CTCCCACAGC TCGT~ACTCT CCCAC~CCC~ A~CCTCCACG 60 ~2) INFOR~AT~ON FOR SEQ ID NO:22;
( i) 513~UENC~: C~ACTERISTICS:
( A ) ~ENGT~ ~ 60 ba~e pair~
3 ) TYPE: nucleic acid C ~ STRAN~5Dr~e~S: ~ ~ ngl e ~ D ) TOPOI,OGY: unknown oL~C~B TYP~: D~ (genc~nlc~
~xi~ SEl~ DR5C~I~TION: SEQ ~D t:O;22:
,,AGAC T~CAGGCC~C CAGTAG~C6 CACGTCGAGA A ~ AC CGTGGAGGTT 60 AMEN~EO S~F~T

Claims (39)

WHAT IS CLAIMED IS:
1. A recombinant adenovirus comprising a chimeric penton base protein, wherein said chimeric penton base protein comprises a nonpenton base amino acid sequence, which is (i) specific for binding to a receptor, (ii) an antibody domain or (iii) an epitope, in addition to or in place of a wild-type penton base amino acid sequence.
2. The recombinant adenovirus of claim 1, where said wild-type penton base amino acid sequence is an RGD
amino acid sequence.
3. The recombinant adenovirus of claim 1, wherein said recombinant adenovirus is fiber-deficient.
4. The recombinant adenovirus of claim 2, which further comprises, in addition to the chimeric penton base protein, a chimeric coat protein which includes an amino acid sequence that is (i) pecific for binding to a receptor, (ii) an antibody domain or (iii) an epitope.
5. The recombinant adenovirus of claim 4, wherein said amino acid sequence in said chimeric coat protein, which is (i) specific for binding to a receptor, (ii) an antibody domain or (iii) an epitope, (i) is specific for binding to the same receptor, (ii) is the same antibody domain or (iii) is the same epitope as the nonpenton base amino acid sequence in said chimeric penton base protein.
6. The recombinant adenovirus of claim 4, wherein aid chimeric coat protein is selected from the group consisting of a chimeric fiber and a chimeric hexon.
7. The recombinant adenovirus of claim 6, wherein said amino acid sequence in said chimeric coat protein, which is (i) specific for binding to a receptor, (ii) an antibody domain or (iii) an epitope, (i) is specific for binding to the same receptor, (ii) is the same antibody domain or (iii) is the same epitope as the nonpenton base amino acid sequence in said chimeric penton base protein.
8. A recombinant baculovirus comprising a chimeric adenoviral penton base gene sequence encoding a chimeric penton base protein, which chimeric adenoviral penton base gene sequence comprises (a) a nonpenton base gene sequence encoding an amino acid sequence that is (i) specific for binding to a receptor, (ii) an antibody domain or (iii) an epitope, and (b) a wild-type penton base gene sequence.
9. The recombinant baculovirus of claim 8, wherein said wild-type penton base gene sequence encodes an RGD
amino acid sequence.
10. The recombinant baculovirus of claim 9, wherein the gene sequence encoding the RGD amino acid sequence has been mutated.
11. The recombinant baculovirus of claim 10, wherein the gene sequence encoding the RGD amino acid sequence has been mutated by insertion of the nonpenton base gene sequence into the gene sequence encoding the RGD amino acid sequence.
12. The recombinant baculovirus of claim 9, wherein the gene sequence encoding the RGD amino acid sequence has been moved within the penton base gene sequence such that in the chimeric penton base protein the RGD amino acid sequence is conformationally inaccessible for binding to a receptor.
13. The recombinant baculovirus of claim 12, wherein the gene sequence encoding the RGD amino acid sequence has been moved within the penton base gene sequence by insertion of the nonpenton base gene sequence near the gene sequence encoding the RGD amino acid.
14. A recombinant baculovirus comprising a chimeric adenoviral penton base gene sequence, wherein said chimeric adenoviral penton base gene sequence includes a nonpenton base gene sequence, which encodes an amino acid sequence that is (i) specific for binding to a receptor, (ii) an antibody domain or (iii) an epitope, in place of a wild-type penton base gene sequence.
15. The recombinant baculovirus of claim 14, wherein said wild-type penton base gene sequence encodes an RGD amino acid sequence.
16. A chimeric adenoviral penton base protein comprising a nonpenton base amino acid sequence, which is (i) specific for binding to a receptor, (ii) an antibody domain or (iii) an epitope, in addition to or in place of a wild-type penton base amino acid sequence.
17. The chimeric adenoviral penton base protein of claim 16, wherein said wild-type penton base amino acid sequence is an RGD amino acid sequence.
18, A viral transfer vector comprising a chimeric adenoviral penton base gene sequence encoding a chimeric penton base protein, which chimeric adenoviral penton base gene sequence comprises a nonpenton base gene sequence encoding an amino acid sequence that is (i) specific for binding to a receptor, (ii) an antibody domain or (iii) an epitope, in addition to or in place of a wild-type penton base gene sequence.
19. The viral transfer vector of claim 19, wherein said viral transfer vector is an adenoviral transfer vector.
20. The viral transfer vector of claim 19, wherein said wild-type penton base gene sequence encodes an RGD
amino acid sequence.
21. The viral transfer vector of claim 20, wherein the gene sequence encoding the RGD amino acid sequence has been mutated.
22. The viral transfer vector of claim 21, wherein the gene sequence encoding the RGD amino acid sequence has been mutated by insertion of the nonpenton base gene sequence into the gene sequence encoding the RGD amino acid sequence.
23. The viral transfer vector of claim 20, wherein the gene sequence encoding the RGD amino acid sequence has been moved within the penton base gene sequence such that in the chimeric penton base protein the RGD amino acid sequence is conformationally inaccessible for binding to a receptor.
24. The viral transfer vector of claim 23, wherein the gene sequence encoding the RGD amino acid sequence has been moved by insertion of the nonpenton base gene sequence near the gene sequence encoding the RGD amino acid sequence,
25. A prokaryotic or eukaryotic expression vector comprising a chimeric adenoviral penton base gene sequence encoding a chimeric penton base protein, which chimeric adenoviral penton base gene sequence comprises a nonpenton base gene sequence encoding an amino acid sequence that is (i) specific for binding to a receptor, (ii) an antibody domain or (iii) an epitope, in addition to or in place of a wild-type penton base gene sequence.
26. The prokaryotic or eukaryotic expression vector of claim 25, wherein said wild-type penton base gene sequence encodes an RGD amino acid sequence.
27. The prokaryotic or eukaryotic expression vector of claim 26, wherein the gene sequence encoding the RGD
amino acid sequence has been mutated.
28. The prokaryotic or eukaryotic expression vector of claim 27, wherein the gene sequence encoding the RGD
amino acid sequence has been mutated by insertion of the nonpenton base gene sequence into the gene sequence encoding the RGD amino acid sequence.
29. The prokaryotic or eukaryotic expression vector of claim 26, wherein the gene sequence encoding the RGD
amino acid sequence has been moved within the penton base gene sequence such that in the chimeric penton base protein the RGD amino acid sequence is conformationally inaccessible for binding to a receptor.
30. The prokaryotic or eukaryotic expression vector of claim 29, wherein the gene sequence encoding the RGD
amino acid sequence has been moved by insertion of the nonpentan base gene sequence near the gene sequence encoding the RGD amino acid sequence.
31. The recombinant adenovirus of any of claims 1-7, wherein said adenovirus further comprises a therapeutic gene that is capable of being expressed in a cell to which the recombinant adenovirus has attached or by which the recombinant adenovirus has been internalized.
32. The recombinant adenovirus of claim 2, wherein said wild-type penton base amino acid sequence is specific for binding to a receptor,
33, The recombinant baculovirus of claim 8, wherein said wild-type penton base gene sequence encodes an amino acid sequence that is specific for binding to a receptor.
34, The recombinant baculovirus of claim 14, wherein said wild-type penton base gene sequence encodes an amino acid sequence that is specific for binding to a receptor.
35. The chimeric adenoviral penton base protein of claim 16, wherein said wild-type penton base amino acid sequence is specific for binding to a receptor.
36. The viral transfer vector of claim 18, wherein said wild-type penton base gene sequence encodes an amino acid sequence that is specific for binding to a receptor.
37. The prokaryotic or eukaryotic expression vector of claim 25, wherein said wild-type penton base gene sequence encodes an amino acid sequence that is specific for binding to a receptor.
38. A method of gene therapy comprising administering to a patient in need of gene therapy a therapeutically effective amount of a recombinant adenovirus of any of claims 1-7, 31, and 32.
39. The recombinant adenovirus of any of claim 1- 7, 31, and 32 for use in preparing a medicament.
CA002198861A 1994-09-08 1995-08-07 Recombinant adenovirus comprising a chimeric penton base protein Abandoned CA2198861A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/303,162 US5559099A (en) 1994-09-08 1994-09-08 Penton base protein and methods of using same
US303,162 1994-09-08

Publications (1)

Publication Number Publication Date
CA2198861A1 true CA2198861A1 (en) 1996-03-14

Family

ID=23170789

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002198861A Abandoned CA2198861A1 (en) 1994-09-08 1995-08-07 Recombinant adenovirus comprising a chimeric penton base protein

Country Status (8)

Country Link
US (3) US5559099A (en)
EP (2) EP0778889B1 (en)
JP (1) JPH10505242A (en)
AT (1) ATE332363T1 (en)
AU (1) AU684220B2 (en)
CA (1) CA2198861A1 (en)
DE (1) DE69535103D1 (en)
WO (1) WO1996007734A2 (en)

Families Citing this family (178)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9223084D0 (en) * 1992-11-04 1992-12-16 Imp Cancer Res Tech Compounds to target cells
US5543328A (en) * 1993-08-13 1996-08-06 Genetic Therapy, Inc. Adenoviruses having modified fiber proteins
GB9412844D0 (en) * 1994-06-27 1994-08-17 Medical Res Council Improvements in or relating to therapeutic methods
US5965541A (en) * 1995-11-28 1999-10-12 Genvec, Inc. Vectors and methods for gene transfer to cells
US5962311A (en) * 1994-09-08 1999-10-05 Genvec, Inc. Short-shafted adenoviral fiber and its use
US5559099A (en) * 1994-09-08 1996-09-24 Genvec, Inc. Penton base protein and methods of using same
US6465253B1 (en) * 1994-09-08 2002-10-15 Genvec, Inc. Vectors and methods for gene transfer to cells
US5846782A (en) 1995-11-28 1998-12-08 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
US5770442A (en) * 1995-02-21 1998-06-23 Cornell Research Foundation, Inc. Chimeric adenoviral fiber 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
US6783980B2 (en) * 1995-06-15 2004-08-31 Crucell Holland B.V. Packaging systems for human recombinant adenovirus to be used in gene therapy
DE69633565T3 (en) 1995-06-15 2013-01-17 Crucell Holland B.V. PACKAGING SYSTEMS FOR HUMAN, HUMAN ADENOVIRES, FOR USE IN GENE THERAPY
CA2224907A1 (en) * 1995-07-25 1997-02-13 Introgene B.V. Methods and means for targeted gene delivery
ATE382694T1 (en) * 1995-11-28 2008-01-15 Genvec Inc VECTORS AND METHODS OF GENE TRANSFER INTO CELLS
US6136311A (en) 1996-05-06 2000-10-24 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
FR2751343B1 (en) * 1996-07-16 1998-12-18 Transgene Sa PROCESS FOR THE PRESERVATION OF INFECTIOUS RECOMBINANT VIRUSES, AQUEOUS VIRAL SUSPENSION, AND USE AS A MEDICAMENT
US7232899B2 (en) * 1996-09-25 2007-06-19 The Scripps Research Institute Adenovirus vectors, packaging cell lines, compositions, and methods for preparation and use
NZ334827A (en) * 1996-09-25 2001-02-23 Scripps Research Inst High-capacity adenoviral vectors suitable for adenoviral-based gene therapy
FR2756297B1 (en) * 1996-11-22 1999-01-08 Centre Nat Rech Scient PROCESS FOR PRODUCING RECOMBINANT VIRUSES
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
US6218597B1 (en) 1997-04-03 2001-04-17 University Technology Corporation Transgenic model and treatment for heart disease
IL133010A0 (en) * 1997-05-28 2001-03-19 Genvec Inc Alternatively targeted adenovirus
US7276488B2 (en) * 1997-06-04 2007-10-02 Oxford Biomedica (Uk) Limited Vector system
WO1998055607A2 (en) * 1997-06-04 1998-12-10 Oxford Biomedica (Uk) Limited Tumor targeted vector
WO2001036486A2 (en) * 1999-11-18 2001-05-25 Oxford Biomedica (Uk) Limited Scfv antibodies against disease associated molecules
US7635687B2 (en) * 1997-06-04 2009-12-22 Oxford Biomedica (Uk) Limited Vector system
CA2321135A1 (en) * 1998-02-17 1999-08-19 Uab Research Foundation Modified adenovirus containing a fiber replacement protein
AU3358999A (en) * 1998-03-20 1999-10-11 Genzyme Corporation Chimeric adenoviral vectors for targeted gene delivery
WO1999055365A1 (en) 1998-04-30 1999-11-04 Cornell Research Foundation, Inc. Adenoviral vectors with tandem fiber proteins
AU4089899A (en) 1998-05-20 1999-12-06 University Of Tennessee Research Corporation, The Stable envelope proteins for retroviral, viral and liposome vectors and use in gene and drug therapy
US20040043489A1 (en) * 1998-07-08 2004-03-04 Menzo Havenga Gene delivery vectors provided with a tissue tropism for dendritic cells and methods of use
US20030017138A1 (en) * 1998-07-08 2003-01-23 Menzo Havenga Chimeric adenoviruses
WO2000015823A1 (en) 1998-09-11 2000-03-23 Genvec, Inc. Alternatively targeted adenovirus
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
AU2869300A (en) * 1999-02-05 2000-08-25 Uab Research Foundation, The Fiber receptor-independent system for the propagation of adenoviral vectors
US6492169B1 (en) * 1999-05-18 2002-12-10 Crucell Holland, B.V. Complementing cell lines
US7468181B2 (en) * 2002-04-25 2008-12-23 Crucell Holland B.V. Means and methods for the production of adenovirus vectors
US20050232900A1 (en) * 1999-05-18 2005-10-20 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US6913922B1 (en) * 1999-05-18 2005-07-05 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US7261881B1 (en) 1999-05-20 2007-08-28 Yale University Modulation of angiogenesis and wound healing
WO2001016344A1 (en) * 1999-08-27 2001-03-08 Transgene S.A. Modified adenoviral fibre and uses thereof
ES2183752T3 (en) * 1999-09-17 2005-07-16 Tgt Laboratories, S.A. De C.V. RECOMBINANT ADENOVIRAL VECTORS AND THEIR USE IN THE TREATMENT OF THE LIVER CIRROSIS.
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
WO2001092549A2 (en) * 2000-05-31 2001-12-06 Genvec, Inc. Method and composition for targeting an adenoviral vector
WO2001092547A2 (en) * 2000-05-31 2001-12-06 University Of Saskatchewan Modified bovine adenovirus having altered tropism
US7355019B2 (en) * 2000-06-06 2008-04-08 Sibtech, Inc. Cysteine-containing peptide tag for site-specific conjugation of proteins
US20020164333A1 (en) * 2000-07-10 2002-11-07 The Scripps Research Institute Bifunctional molecules and vectors complexed therewith for targeted gene delivery
EP1322774A2 (en) * 2000-09-20 2003-07-02 Crucell Holland B.V. Gene delivery vectors provided with a tissue tropism for dendritic cells
DE60138403D1 (en) * 2000-09-26 2009-05-28 Crucell Holland Bv ADENOVIRAL VECTORS FOR THE TRANSFER OF GENES IN CELLS OF THE SKELETAL MUSCULATORY OR MYOBLAST
US6573092B1 (en) * 2000-10-10 2003-06-03 Genvec, Inc. Method of preparing a eukaryotic viral vector
US20030202963A1 (en) * 2000-10-12 2003-10-30 Cornell Research Foundation, Inc. Method of treating cancer
MXPA00011713A (en) * 2000-11-28 2002-05-31 Tgt Lab S A De C V Re-combining viral and non-viral vectors containing the human gene of the urokinase plasminogen activator and its benefit in the treatment of diverse types of hepatic, renal, pulmonary, pancreatic and cardiac fibrosis and hypertrophic scars.
EP1390476A4 (en) * 2001-04-17 2005-03-30 Vectorlogics Inc Mosaic adenoviral vectors
US20060275262A1 (en) * 2001-07-26 2006-12-07 Mathis James M Conditionally replicating viruses and methods for cancer virotherapy
GB0119852D0 (en) * 2001-08-15 2001-10-10 Univ York Baculovirus
WO2003018752A2 (en) * 2001-08-23 2003-03-06 The Wistar Institute Of Anatomy And Biology An organotypic intestinal culture and methods of use thereof
US7247297B2 (en) * 2001-09-14 2007-07-24 The University Of Chicago Use of DF3/MUC1 regulated expression in gene therapy
US20040031067A1 (en) * 2001-10-11 2004-02-12 Meenhard Herlyn Regulation of human skin healing
GB0126378D0 (en) * 2001-11-02 2002-01-02 Oxford Biomedica Ltd Antigen
EP1516055A4 (en) * 2002-01-24 2007-08-08 Scripps Research Inst Fiber shaft modifications for efficient targeting
AU2003223775A1 (en) * 2002-04-30 2003-11-17 Duke University Adeno-associated viral vectors and methods for their production from hybrid adenovirus and for their use
CA2495546A1 (en) * 2002-08-22 2004-03-04 Merck & Co., Inc. Methods for propagating adenovirus and virus produced thereby
US9701754B1 (en) 2002-10-23 2017-07-11 City Of Hope Covalent disulfide-linked diabodies and uses thereof
ATE521701T1 (en) 2003-01-22 2011-09-15 Univ Duke IMPROVED CONSTRUCTS FOR EXPRESSING LYSOSOMAL POLYPEPTIDES
US20040166091A1 (en) * 2003-02-24 2004-08-26 Genvec, Inc. Materials and methods for treating disorders of the ear
WO2004092397A2 (en) * 2003-04-15 2004-10-28 Novartis Ag Tmprss2 regulatory sequences and uses thereof
WO2004092396A2 (en) * 2003-04-15 2004-10-28 Novartis Ag Flap endonuclease 1 (fen1) regulatory sequences and uses thereof
EP1631582A2 (en) * 2003-06-10 2006-03-08 University of Saskatchewan Chimeric adenovirus capsid proteins
US7291498B2 (en) * 2003-06-20 2007-11-06 The Trustees Of The University Of Pennsylvania Methods of generating chimeric adenoviruses and uses for such chimeric adenoviruses
US7491508B2 (en) * 2003-06-20 2009-02-17 The Trustees Of The University Of Pennsylvania Methods of generating chimeric adenoviruses and uses for such chimeric adenoviruses
ES2391975T3 (en) * 2003-07-25 2012-12-03 Genvec, Inc. Adenoviral vector based vaccines
US7519698B2 (en) * 2003-09-26 2009-04-14 Ricoh Co., Ltd. Method and system for extracting information from networked devices in a multi-protocol remote monitoring system
WO2005051432A1 (en) * 2003-11-14 2005-06-09 Genvec, Inc. Therapeutic regimen for treating cancer
ES2329607T3 (en) * 2004-02-23 2009-11-27 Crucell Holland B.V. VIRUS PURIFICATION METHODS
US7473418B2 (en) * 2004-03-25 2009-01-06 Cell Genesys, Inc. Pan cancer oncolytic vectors and methods of use thereof
CN1972958B (en) 2004-04-12 2013-01-23 美国政府卫生与公共服务部 Method of using adenoviral vectors to induce an immune response
US20060062764A1 (en) * 2004-08-25 2006-03-23 Seshidar-Reddy Police Fiber-modified adenoviral vectors for enhanced transduction of tumor cells
WO2006039045A2 (en) * 2004-09-01 2006-04-13 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Adenoviral vectors able to transduce apcs, potential use in immune response generation
US20070275874A1 (en) * 2004-09-03 2007-11-29 Yale University Use of Leptin in Wound Healing
US20060205080A1 (en) * 2005-03-01 2006-09-14 David Frey Formulations for therapeutic viruses having enhanced storage stability
CA2602944C (en) * 2005-04-11 2015-08-11 Crucell Holland B.V. Virus purification using ultrafiltration
US20060234347A1 (en) * 2005-04-13 2006-10-19 Harding Thomas C Targeting multiple angiogenic pathways for cancer therapy using soluble tyrosine kinase receptors
US8450055B2 (en) * 2005-08-31 2013-05-28 The United States Of America As Represented By The Secretary Of The Navy Malaria antigen screening method
EP1929021A2 (en) * 2005-08-31 2008-06-11 Genvec, Inc. Adenoviral vector-based malaria vaccines
US9651543B2 (en) 2005-08-31 2017-05-16 The United States Of America As Represented By The Secretary Of The Navy Malaria antigen screening method
BRPI0618441B8 (en) * 2005-11-10 2021-07-27 Genvec Inc adenoviral vector
EP1996716B1 (en) 2006-03-20 2011-05-11 The Regents of the University of California Engineered anti-prostate stem cell antigen (psca) antibodies for cancer targeting
WO2007123737A2 (en) 2006-03-30 2007-11-01 University Of California Methods and compositions for localized secretion of anti-ctla-4 antibodies
US7919079B2 (en) * 2006-03-31 2011-04-05 Biosante Pharmaceuticals, Inc. Cancer immunotherapy compositions and methods of use
US20100278870A1 (en) * 2007-01-09 2010-11-04 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Adenoviral vector-based malaria vaccines
WO2009009441A2 (en) 2007-07-06 2009-01-15 Cedars-Sinai Medical Center Self-assembling complex for targeting chemical agents to cells
JP6126773B2 (en) 2007-09-04 2017-05-10 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア High affinity anti-prostatic stem cell antigen (PSCA) antibody for cancer targeting and detection
EP2209498A2 (en) * 2007-10-03 2010-07-28 Cornell University Treatment of proliferative disorders using antibodies to psma
US20090202492A1 (en) * 2008-01-25 2009-08-13 University Of South Florida Adenovirus vaccine utilizing ikk as adjuvant
US8709316B2 (en) * 2008-03-14 2014-04-29 Dow Global Technologies Llc Process for shaping polymeric articles
US20100069616A1 (en) * 2008-08-06 2010-03-18 The Regents Of The University Of California Engineered antibody-nanoparticle conjugates
WO2010060719A1 (en) * 2008-11-03 2010-06-03 Crucell Holland B.V. Method for the production of adenoviral vectors
WO2010085665A2 (en) 2009-01-23 2010-07-29 Cedars-Sinai Medical Center Targeted delivery system
WO2010096486A1 (en) * 2009-02-17 2010-08-26 Cornell Research Foundation, Inc. Methods and kits for diagnosis of cancer and prediction of therapeutic value
US9605844B2 (en) * 2009-09-01 2017-03-28 Cree, Inc. Lighting device with heat dissipation elements
US8470585B2 (en) 2009-10-15 2013-06-25 Crucell Holland B.V. Process for adenovirus purification from high cell density cultures
JP5393896B2 (en) 2009-10-15 2014-01-22 クルセル ホランド ベー ヴェー Method for purifying adenovirus particles
WO2011047316A1 (en) 2009-10-16 2011-04-21 The United States Of America, As Represented By The Secretary Department Of Health And Human Services Nucleic acid sequences encoding expandable hiv mosaic proteins
WO2011057254A2 (en) 2009-11-09 2011-05-12 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Simian adenoviral vector-based vaccines
AU2010314842A1 (en) 2009-11-09 2012-05-31 Genvec, Inc. Methods of propagating monkey adenoviral vectors
CA2782333C (en) 2009-12-02 2019-06-04 Imaginab, Inc. J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use
PL2536829T3 (en) 2010-02-15 2016-09-30 Method for the production of Ad26 adenoviral vectors
US9682133B2 (en) 2010-03-17 2017-06-20 Cornell University Disrupted adenovirus-based vaccine against drugs of abuse
TWI688395B (en) 2010-03-23 2020-03-21 英翠克頌公司 Vectors conditionally expressing therapeutic proteins, host cells comprising the vectors, and uses thereof
WO2012021730A2 (en) 2010-08-11 2012-02-16 Genvec, Inc. Respiratory syncytial virus (rsv) vaccine
CN103118702A (en) 2010-09-20 2013-05-22 克鲁塞尔荷兰公司 Therapeutic vaccination against active tuberculosis
MX354752B (en) 2010-09-27 2018-03-20 Janssen Vaccines & Prevention Bv Heterologous prime boost vaccination regimen against malaria.
US9512429B2 (en) 2010-12-06 2016-12-06 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Pharmaceutical composition comprising Nanog shRNA, and method of using Nanog shRNA to treat cancer
WO2012083297A2 (en) 2010-12-17 2012-06-21 Genvec, Inc. Adenoviral vectors with modified hexon regions
EP2654786B1 (en) 2010-12-20 2019-02-20 GenVec, Inc. Adenoviral vector-based dengue fever vaccine
WO2012122025A2 (en) 2011-03-04 2012-09-13 Intrexon Corporation Vectors conditionally expressing protein
US20130122038A1 (en) 2011-11-14 2013-05-16 The United States Of America As Represented By The Secretary Of The Department Heterologous prime-boost immunization using measles virus-based vaccines
US8932607B2 (en) 2012-03-12 2015-01-13 Crucell Holland B.V. Batches of recombinant adenovirus with altered terminal ends
WO2013135615A1 (en) 2012-03-12 2013-09-19 Crucell Holland B.V. Batches of recombinant adenovirus with altered terminal ends
US9119813B2 (en) 2012-03-22 2015-09-01 Crucell Holland B.V. Vaccine against RSV
MY169331A (en) 2012-03-22 2019-03-21 Janssen Vaccines & Prevention Bv Vaccine against rsv
EP2855669B1 (en) 2012-05-29 2018-10-10 GenVec, Inc. Modified serotype 28 adenoviral vectors
ES2703052T3 (en) 2012-08-03 2019-03-06 Cedars Sinai Medical Center Isolation of protein-enhancing drug delivery mutants
KR102089121B1 (en) 2013-03-14 2020-03-13 더 솔크 인스티튜트 포 바이올로지칼 스터디즈 Oncolytic adenovirus compositions
JP6576904B2 (en) 2013-04-04 2019-09-18 トラスティーズ・オブ・ダートマス・カレッジ Compositions and methods for in vivo excision of HIV-1 proviral DNA
KR102236497B1 (en) 2013-04-25 2021-04-06 얀센 백신스 앤드 프리벤션 비.브이. Stabilized soluble prefusion rsv f polypeptides
PL2993983T3 (en) 2013-05-08 2020-09-07 Cedars-Sinai Medical Center Targeting corroles for tumor toxicity and mri
US10294279B2 (en) 2013-06-17 2019-05-21 Janssen Vaccines & Prevention B.V. Stabilized soluble pre-fusion RSV F polypeptides
AP2016009154A0 (en) 2013-09-19 2016-04-30 Crucell Holland Bv Improved adenovirus formulations
JP6608370B2 (en) 2014-01-17 2019-11-20 シーダーズ―シナイ メディカル センター Receptor targeting constructs and uses thereof
WO2015154059A2 (en) 2014-04-04 2015-10-08 Cedars-Sinai Medical Center Targeting trastuzumab-resistant her2+breast cancer with a her3-targeting nanoparticle
TWI710635B (en) 2014-10-09 2020-11-21 美商珍維克公司 Adenoviral vector encoding human atonal homolog-1 (hath1)
RS58080B1 (en) 2014-11-04 2019-02-28 Janssen Vaccines & Prevention Bv Therapeutic hpv16 vaccines
JP2018504141A (en) 2015-01-20 2018-02-15 アドキュア バイオテクノロジーズ、 エルエルシー Non-targeted adenovirus variants and related methods
JP6887955B2 (en) 2015-03-18 2021-06-16 ヤンセン ファッシンズ アンド プリベンション ベーフェーJanssen Vaccines & Prevention B.V. Recombinant expression system assay
AU2016249798B2 (en) 2015-04-14 2022-05-26 Janssen Vaccines And Prevention B.V. Recombinant adenovirus expressing two transgenes with a bidirectional promoter
PL3319633T3 (en) 2015-07-07 2021-04-19 Janssen Vaccines & Prevention B.V. Vaccine against rsv
EP3319634B1 (en) 2015-07-07 2019-08-21 Janssen Vaccines & Prevention B.V. Stabilized soluble pre-fusion rsv f polypeptides
NZ739721A (en) 2015-08-07 2019-09-27 Imaginab Inc Antigen binding constructs to target molecules
AU2016309743B2 (en) 2015-08-20 2019-02-07 Janssen Vaccines & Prevention B.V. Therapeutic HPV18 vaccines
WO2017060329A1 (en) 2015-10-06 2017-04-13 Janssen Vaccines & Prevention B.V. Methods for preventing plastic-induced degradation of biologicals
WO2017070113A1 (en) 2015-10-19 2017-04-27 The United States of America, as represented by the Secretary, Department of Health and Human Sevices Pharmaceutical composition comprising nanog shrna, and method of using nanog shrna to treat cancer
AU2017222568B2 (en) 2016-02-23 2020-09-10 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
KR20220163505A (en) 2016-02-23 2022-12-09 솔크 인스티튜트 포 바이올로지칼 스터디즈 Exogenous gene expression in therapeutic adenovirus for minimal impact on viral kinetics
JP7233928B2 (en) 2016-04-05 2023-03-07 ヤンセン ファッシンズ アンド プリベンション ベーフェー Vaccine against RSV
SI3439672T1 (en) 2016-04-05 2021-02-26 Janssen Vaccines & Prevention B.V. Stabilized soluble pre-fusion rsv f protein for use in the prophylaxis of rsv infection
WO2017192418A1 (en) 2016-05-02 2017-11-09 Janssen Vaccine & Prevention B.V. Therapeutic hpv vaccine combinations
CN109154000B (en) 2016-05-12 2022-07-05 扬森疫苗与预防公司 Efficient and balanced bidirectional promoters
EA039124B1 (en) 2016-05-30 2021-12-08 Янссен Вэксинс Энд Превеншн Б.В. Stabilized pre-fusion rsv f proteins
EP3472327B1 (en) 2016-06-20 2020-08-19 Janssen Vaccines & Prevention B.V. Potent and balanced bidirectional promoter
EP3484506A1 (en) 2016-07-14 2019-05-22 Janssen Vaccines & Prevention B.V. Hpv vaccines
US11155832B2 (en) 2016-09-30 2021-10-26 Genvec, Inc. Adenovectors for delivery of therapeutic genetic material into T cells
AU2017375633C1 (en) 2016-12-12 2023-04-27 Salk Institute For Biological Studies Tumor-targeting synthetic adenoviruses and uses thereof
WO2018147960A1 (en) 2017-02-08 2018-08-16 Imaginab, Inc. Extension sequences for diabodies
BR112019015671A2 (en) 2017-02-09 2020-04-14 Janssen Vaccines & Prevention Bv potent and short promoter for expression of heterological genes
CA3061278A1 (en) 2017-05-17 2018-11-22 Janssen Vaccines & Prevention B.V. Methods and compositions for inducing protective immunity against rsv infection
AU2018333566A1 (en) 2017-09-15 2020-02-27 Janssen Vaccines & Prevention B.V. Method for the safe induction of immunity against RSV
US11459583B2 (en) 2017-10-31 2022-10-04 Janssen Vaccines & Prevention B.V. Adenovirus vectors and uses thereof
MA50502A (en) 2017-10-31 2020-09-09 Janssen Vaccines & Prevention Bv ADENOVIRUS AND RELATED USES
CN111295391B (en) 2017-10-31 2023-12-05 扬森疫苗与预防公司 Adenovirus and use thereof
JP7438943B2 (en) 2017-10-31 2024-02-27 ヤンセン ファッシンズ アンド プリベンション ベーフェー Adenovirus and its uses
PT3743106T (en) 2018-01-23 2022-08-24 Janssen Vaccines & Prevention Bv Influenza virus vaccines and uses thereof
TW202043256A (en) 2019-01-10 2020-12-01 美商健生生物科技公司 Prostate neoantigens and their uses
WO2020172509A1 (en) 2019-02-21 2020-08-27 Unleash Immuno Oncolytics, Inc. Oncolytic adenoviral vector and methods of use
WO2020216844A1 (en) 2019-04-25 2020-10-29 Janssen Vaccines & Prevention B.V. Recombinant influenza antigens
CA3140234A1 (en) 2019-05-15 2020-11-19 Janssen Vaccines & Prevention B.V. Prophylactic treatment of respiratory syncytial virus infection with an adenovirus based vaccine
AU2020275455A1 (en) 2019-05-15 2021-12-09 Janssen Vaccines & Prevention B.V. Co-administration of seasonal influenza vaccine and an adenovirus based respiratory syncytial virus vaccine
JP2022547107A (en) 2019-09-05 2022-11-10 ヤンセン ファッシンズ アンド プリベンション ベーフェー Influenza virus vaccine and its use
JP2022551107A (en) 2019-10-03 2022-12-07 ヤンセン ファッシンズ アンド プリベンション ベーフェー Adenoviral vectors and uses thereof
IL293051A (en) 2019-11-18 2022-07-01 Janssen Biotech Inc Vaccines based on mutant calr and jak2 and their uses
TW202144388A (en) 2020-02-14 2021-12-01 美商健生生物科技公司 Neoantigens expressed in ovarian cancer and their uses
TW202144389A (en) 2020-02-14 2021-12-01 美商健生生物科技公司 Neoantigens expressed in multiple myeloma and their uses
AU2021357520A1 (en) 2020-03-05 2022-09-29 Neotx Therapeutics Ltd. Methods and compositions for treating cancer with immune cells
EP4135757A1 (en) 2020-04-13 2023-02-22 Janssen Biotech, Inc. Psma and steap1 vaccines and their uses
WO2022009051A1 (en) 2020-07-06 2022-01-13 Janssen Biotech, Inc. A method for determining responsiveness to prostate cancer treatment
US20230024133A1 (en) 2020-07-06 2023-01-26 Janssen Biotech, Inc. Prostate Neoantigens And Their Uses
WO2022009052A2 (en) 2020-07-06 2022-01-13 Janssen Biotech, Inc. Prostate neoantigens and their uses
EP4338727A1 (en) 2022-09-14 2024-03-20 Roquette Freres Adenovirus formulations

Family Cites Families (27)

* 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
US4578079A (en) * 1982-08-04 1986-03-25 La Jolla Cancer Research Foundation Tetrapeptide
US4792525A (en) * 1982-08-04 1988-12-20 La Jolla Cancer Research Foundation Tetrapeptide
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
CA2000048A1 (en) * 1988-10-03 1990-04-03 Edward F. Plow Peptides and antibodies that inhibit integrin-ligand bindin g
DE69029036T2 (en) * 1989-06-29 1997-05-22 Medarex Inc SPECIFIC REAGENTS FOR AIDS THERAPY
US5332567A (en) * 1989-08-24 1994-07-26 Immunomedics Detection and treatment of infections with immunoconjugates
CA2067244C (en) * 1989-10-20 2000-12-19 Li Shen Monoclonal antibody specific for iga receptor
ATE177152T1 (en) * 1989-10-20 1999-03-15 Medarex Inc BISPECIFIC HETEROANTIBODIES WITH DUAL EFFECTOR FUNCTIONS
US5349053A (en) * 1990-06-01 1994-09-20 Protein Design Labs, Inc. Chimeric ligand/immunoglobulin molecules and their uses
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
IL103059A0 (en) * 1991-09-30 1993-02-21 Boehringer Ingelheim Int Conjugates for introducing nucleic acid into higher eucaryotic cells
NZ244306A (en) * 1991-09-30 1995-07-26 Boehringer Ingelheim Int Composition for introducing nucleic acid complexes into eucaryotic cells, complex containing nucleic acid and endosomolytic agent, peptide with endosomolytic domain and nucleic acid binding domain and preparation
US5521291A (en) * 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
GB9223084D0 (en) * 1992-11-04 1992-12-16 Imp Cancer Res Tech Compounds to target cells
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
US5543328A (en) * 1993-08-13 1996-08-06 Genetic Therapy, Inc. Adenoviruses having modified fiber proteins
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.
US5443953A (en) * 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
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
CA2158926A1 (en) * 1994-05-13 1995-11-14 Douglas J. Jolly Compositions and methods for targeting gene delivery vehicles
US5559099A (en) * 1994-09-08 1996-09-24 Genvec, Inc. Penton base protein and methods of using same

Also Published As

Publication number Publication date
ATE332363T1 (en) 2006-07-15
US5712136A (en) 1998-01-27
EP0778889A2 (en) 1997-06-18
AU684220B2 (en) 1997-12-04
AU3212995A (en) 1996-03-27
US5731190A (en) 1998-03-24
DE69535103D1 (en) 2006-08-17
EP1679376A2 (en) 2006-07-12
JPH10505242A (en) 1998-05-26
WO1996007734A3 (en) 1996-05-09
EP1679376A3 (en) 2006-08-16
US5559099A (en) 1996-09-24
WO1996007734A2 (en) 1996-03-14
EP0778889B1 (en) 2006-07-05

Similar Documents

Publication Publication Date Title
CA2198861A1 (en) Recombinant adenovirus comprising a chimeric penton base protein
AU698254B2 (en) Chimeric adenoviral fiber protein and methods of using same
AU734515B2 (en) A short-shafted adenoviral fiber and its use
ES2246533T3 (en) MODIFIED ADENOVIRICAL FIBER AND ADENOVIRUS DIANAS.
AU724189B2 (en) Vectors and methods for gene transfer to cells
US5965541A (en) Vectors and methods for gene transfer to cells
US6057155A (en) Targeting adenovirus with use of constrained peptide motifs
US6455314B1 (en) Alternatively targeted adenovirus
JP2002507391A5 (en)
JP2003508057A (en) Modified adenovirus fibers and uses
JP2003534805A (en) Modified bovine adenovirus with altered affinity

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued