EP1224313A1 - Virus adeno-associe et utilisations correspondantes - Google Patents

Virus adeno-associe et utilisations correspondantes

Info

Publication number
EP1224313A1
EP1224313A1 EP00970689A EP00970689A EP1224313A1 EP 1224313 A1 EP1224313 A1 EP 1224313A1 EP 00970689 A EP00970689 A EP 00970689A EP 00970689 A EP00970689 A EP 00970689A EP 1224313 A1 EP1224313 A1 EP 1224313A1
Authority
EP
European Patent Office
Prior art keywords
dna
aav
recombinant
dna segment
dna molecule
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP00970689A
Other languages
German (de)
English (en)
Inventor
John F. Engelhardt
Dongsheng Duan
Ziyang Yan
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.)
University of Iowa Research Foundation UIRF
Original Assignee
University of Iowa Research Foundation UIRF
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 University of Iowa Research Foundation UIRF filed Critical University of Iowa Research Foundation UIRF
Publication of EP1224313A1 publication Critical patent/EP1224313A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/108Plasmid DNA episomal 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/38Vector systems having a special element relevant for transcription being a stuffer
    • 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
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES
    • 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
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/44Vectors comprising a special translation-regulating system being a specific part of the splice mechanism, e.g. donor, acceptor
    • 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
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/44Vectors comprising a special translation-regulating system being a specific part of the splice mechanism, e.g. donor, acceptor
    • C12N2840/445Vectors comprising a special translation-regulating system being a specific part of the splice mechanism, e.g. donor, acceptor for trans-splicing, e.g. polypyrimidine tract, branch point splicing

Definitions

  • Adeno-associated virus is a non-pathogenic parvovirus with a single-stranded DNA genome of 4680 nucleotides.
  • the genome may be of either plus or minus polarity, and codes for two groups of genes, Rep and Cap (Berns et al., 1990).
  • ITRs Inverted terminal repeats
  • adenovirus or he ⁇ es virus adenovirus or he ⁇ es virus
  • wild-type AAV In the absence of a helper virus, wild-type AAV (wtAAV) establishes a latent, non-productive infection with long-term persistence by integrating into a specific locus on chromosome 19, AAVSl, of the host genome through a Rep-facilitated mechanism (Samulski, 1993; Linden et al., 1996; Kotin et al., 1992).
  • wtAAV wild-type AAV
  • rAAV the mechanism(s) of latent phase persistence of recombinant AAV
  • rAAV integration into the host genome is not site-specific due to deletion of the AAV Rep gene (Ponnazhagan et al., 1997).
  • rAAV has recently been recognized as an extremely attractive vehicle for gene delivery (Muzyczka, 1992).
  • rAAV vectors have been developed by substituting all viral open reading frames with a therapeutic minigene, while retaining the cis elements contained in two inverted terminal repeats (ITRs) (Samulski et al., 1987; Samulski et al., 1989).
  • rAAV genomes can persist as episomes (Flotte et al., 1994; Afione et al., 1996; Duan et al., 1998), or alternatively can integrate randomly into the cellular genome (Berns et al., 1996; McLaughlin et al., 1988; Duan et al., 1997; Fisher-Adams et al., 1996; Kearns et al., 1996; Ponnazhagan et al., 1997).
  • rAAV has been shown to be capable of stable, long- term transgene expression both in vitro and in vivo in a variety of tissues
  • the transduction efficiency of rAAV is markedly variable in different cell types.
  • rAAV has been reported to transduce lung epithelial cells at low levels (Halbert et al., 1997; Duan et al., 1998a), while high level, persistent transgene expression has been demonstrated in muscle, neurons and in other non-dividing cells (Kessler et al., 1996; Fisher et al., 1997; Herzog et al., 1997; Xiao et al., 1996; Kaplitt et al., 1994; Wu et al., 1998; Ali et al., 1996; Bennett et al., 1997 Westfall et al., 1997).
  • tissue-specific differences in rAAV mediated gene transfer may, in part, be due to variable levels of cellular factors affecting AAV infectivity (i.e., receptors and co-receptors such as heparin sulfate proteoglycan, FGFR-1, and V ⁇ 5 integrin) (Summerford et al, 1998; Qing et al., 1999; Summerford et al., 1999) as well as the latent life cycle (i.e., nuclear trafficking of virus and/or the conversion of single stranded genomes to expressible forms) (Qing et al., 1997; Qing et al., 1998).
  • AAV infectivity i.e., receptors and co-receptors such as heparin sulfate proteoglycan, FGFR-1, and V ⁇ 5 integrin
  • latent life cycle i.e., nuclear trafficking of virus and/or the conversion of single stranded genomes to expressible forms
  • Muscle-mediated gene transfer represents a very promising approach for the treatment of hereditary myopathies and several other metabolic disorders. Previous studies have demonstrated remarkably efficient and persistent transgene expression to skeletal muscle in vivo with rAAV vectors. Applications in this model system include the treatment of several inherited disorders such as Factor IX deficiency in hemophilia B and epo deficiencies (Kessler et al., 1996; Herzog et al., 1997).
  • a rAAV vector may not be useful if large regulatory elements are needed to control transgene expression.
  • rAAV vectors that have increased stability and/or persistence in host cells.
  • vectors useful to express large open reading frames there is a need for rAAV vectors that have increased stability and/or persistence in host cells.
  • rAAV rAAV vector
  • ITRs AAV inverted terminal repeats
  • the circular intermediate was isolated from rAAV-infected cells by employing a recombinant AAV "shuttle" vector.
  • the shuttle vector comprises: a) a bacterial origin of replication; b) a marker gene or a selectable gene; c) a 5' ITR; and d) a 3' ITR.
  • the recombinant AAV shuttle vector contains a reporter gene, e.g., a GFP, alkaline phosphatase or ⁇ -galactosidase gene, a selectable marker gene, e.g., an ampicillin-resistance gene, a bacterial origin of replication, a 5' ITR and a 3' ITR.
  • the vector is contacted with eukaryotic cells so as to yield transformed eukaryotic cells.
  • Low molecular weight DNA (“Hirt DNA”) from the transformed eukaryotic cells is isolated.
  • Bacterial cells are contacted with the Hirt DNA so as to yield transformed bacterial cells.
  • bacterial cells which express the marker or selectable gene present in the shuttle vector and which comprise at least a portion of a circular intermediate of adeno-associated virus.
  • circularized intermediates of rAAV impart episomal persistence to linked sequences in Hela cells, fibroblasts and muscle cells.
  • the incorporation of certain AAV sequences, e.g., ITRs, from circular intermediates into a heterologous plasmid conferred a 10-fold increase in the stability of plasmid-based vectors in HeLa cells.
  • therapeutic or prophylactic therapies in which the present vectors are useful include blood disorders (e.g., sickle cell anemia, thalassemias, hemophilias, and Fanconi anemias), neurological disorders, such as Alzheimer's disease and Parkinson's disease, and muscle disorders involving skeletal, cardiac or smooth muscle.
  • blood disorders e.g., sickle cell anemia, thalassemias, hemophilias, and Fanconi anemias
  • neurological disorders such as Alzheimer's disease and Parkinson's disease
  • muscle disorders involving skeletal, cardiac or smooth muscle e.g., Alzheimer's disease and Parkinson's disease.
  • therapeutic genes useful in the vectors of the invention include the ⁇ -globin gene, the ⁇ -globin gene, the cystic fibrosis transmembrane conductance receptor gene (CFTR), the erythropoietin (epo) gene, the Fanconi anemia complementation group, a gene encoding a ribozyme, an antisense gene, a low density lipoprotein (LDL) gene, a tyrosine hydroxylase gene (Parkinson's disease), a glucocerebrosidase gene (Gaucher' s disease), an arylsulfatase A gene (metachromatic leukodystrophies) or genes encoding other polypeptides or proteins.
  • CFTR cystic fibrosis transmembrane conductance receptor gene
  • epo erythropoietin
  • Fanconi anemia complementation group a gene encoding a ribozyme, an antisense gene, a low density
  • a vector of the invention i.e., a plurality of genes may be present in an individual vector.
  • a circular intermediate may be a concatamer, each monomer of that concatamer may comprise a different gene, or a portion thereof.
  • helper- free virus can be prepared (see WO 95/13365) from circular intermediates or vectors of the invention.
  • liposomes, plasmid or virosomes may be employed to deliver a vector of the invention to a host or host cell.
  • the increased persistence of circular intermediates or vectors having one or a plurality of ITRs may be due to the primary and/or secondary structure of the ITRs.
  • the primary structure of a consensus sequence (SEQ ID NO:3) of ITRs formed by the juxtaposition and physical (phosphodiester bond) linkage of ITRs from AAV is shown in Figure 2C.
  • each ITR sequence may be incomplete, i.e., the ITR may be a subunit or portion of the full length ITRs present in the consensus sequence.
  • an isolated DNA segment of the invention is not the 165 bp double DD sequence (SEQ ID NO:7) disclosed in U.S. Patent No. 5,478,745, referred to as a "double sequence".
  • the formation, persistence and/or abundance of molecules having the ITR sequences of the invention may be modulated by helper virus, e.g., adenoviral proteins and/or host cell proteins.
  • helper virus e.g., adenoviral proteins and/or host cell proteins.
  • the circular intermediates or vectors of the invention may be useful to identify and/or isolate proteins that bind to the ITR sequences present in those molecules.
  • the present invention provides an isolated and purified DNA molecule comprising at least one DNA segment, a biologically active subunit or variant thereof, of a circular intermediate of adeno-associated virus, which DNA segment confers increased episomal stability, persistence or abundance of the isolated DNA molecule in a host cell.
  • the DNA molecule comprises at least a portion of a left (5') inverted terminal repeat (ITR) of adeno-associated virus.
  • the DNA molecule comprises at least a portion of a right (3 ')-inverted terminal repeat of adeno-associated virus.
  • the invention also provides a gene transfer vector, comprising: at least one first DNA segment, a biologically active subunit or variant thereof, of a circular intermediate of adeno- associated virus, which DNA segment confers increased episomal stability or persistence of the vector in a host cell; and a second DNA segment comprising a gene.
  • the second DNA segment encodes a therapeutically effective polypeptide.
  • the first DNA segment comprises ITR sequences, preferably at least about 100, more preferably at least about 300, and even more preferably at least about 400, bp of adeno-associated virus sequence.
  • a preferred vector of the invention is a plasmid.
  • the vector of the invention is useful in a method of delivering and/or expressing'a gene in a host cell, to prepare host cells having the vector(s), and in the preparation of compositions comprising such vectors.
  • a recombinant adenovirus helper virus may be employed.
  • the implications of intermolecular recombination of rAAV genomes to form a single circular episome, which may be a circular concatamer comprising at least two different rAAV genomes, is particularly relevant for gene therapy with rAAV.
  • large regulatory elements and genes beyond the packaging capacity of rAAV can be brought together by co-infecting tissue with two independent vectors.
  • enhancers and/or promoters may be introduced into one vector while DNA comprising an open reading frame, e.g., a gene of interest, with or without a minimal promoter, is introduced into a second vector.
  • DNA comprising an open reading frame e.g., a gene of interest, with or without a minimal promoter
  • the transgene cassette size is increased beyond that for a single AAV vector alone and the DNA comprising the opening reading frame is linked to the enhancer and/or promoter.
  • vectors encoding two independent regions of a gene are brought together to form an intact splicing unit by circular concatamerization.
  • a vector comprising an origin of replication and a DNA encoding a protein that binds to the origin and promotes replication and/or maintenance of DNA that is linked to the origin, and a vector comprising a gene of interest are brought together after co-infection to form an autonomously replicating episome comprising the gene.
  • the tibialis muscle of mice was co-infected with rAAV Alkaline phosphatase (Alkphos) and GFP encoding vectors.
  • the GFP shuttle vector also encoded ampicillin resistance and a bacterial origin of replication to allow for bacterial rescue of circular intermediates in Hirt DNA from infected muscle samples.
  • one rAAV vector comprises a first DNA segment comprising a 5' ITR linked to a second DNA segment comprising a promoter operably linked to a third DNA segment comprising a first open reading frame linked to a fourth DNA segment comprising a 3 ' ITR.
  • a second rAAV vector comprises a first DNA segment comprising a 5 ' ITR linked to a second DNA segment comprising a promoter operably linked to a third DNA segment comprising a second open reading frame linked to a fourth DNA segment comprising a 3' ITR.
  • one rAAV vector comprises a first DNA segment comprising a 5' ITR linked to a second DNA segment comprising a promoter operably linked to a third DNA segment comprising the 5 ' end of an open reading frame linked to fourth DNA segment comprising a 5' splice site linked to a fifth DNA segment comprising a 3 ' ITR.
  • the second rAAV vector comprises a first DNA segment comprising a 5' ITR linked to a second DNA segment comprising a 3' splice site linked to a third DNA segment comprising the 3' end of the open reading frame linked to a fourth DNA segment comprising a 3 ' ITR.
  • the second and third DNA segments together comprise DNA encoding, for example, CFTR, factor VIII, dystrophin, or erythropoietin.
  • the second DNA segment comprises the endogenous promoter of the respective gene, e.g., the epo promoter.
  • the invention provides a composition comprising: a first adeno- associated virus vector comprising linked DNA segments and at least a second adeno-associated virus comprising linked DNA segments.
  • the linked DNA segments of the first vector comprise: a first DNA segment comprising a 5' ITR; a second DNA segment comprising at least a portion of an open reading frame operably linked to a promoter, wherein the DNA segment does not comprise the entire open reading frame; a third DNA segment comprising a splice donor site; and iv) a fourth DNA segment comprising a 3 ' ITR.
  • the linked DNA segments of the second vector comprise a first DNA segment comprising a 5' ITR; a second DNA segment comprising a splice acceptor site; a third DNA segment comprising at least a portion of an open reading frame which together with the second DNA segment of the first vector encodes a full-length polypeptide; and a fourth DNA segment comprising a 3 ' ITR.
  • the second DNA segment of the first vector comprises a first exon of a gene comprising more than one exon and the third DNA segment of the second vector comprises at least one exon of a gene that is not the first exon.
  • the invention also provides a method to transfer and express a polypeptide in a host cell.
  • the method comprises contacting the host cell with at least two rAAV vectors.
  • One rAAV vector comprises a first DNA segment comprising a 5 'ITR linked to a second DNA segment comprising a promoter operably linked to a third DNA segment comprising a first open reading frame linked to a fourth DNA segment comprising a 3 ' ITR.
  • a second rAAV vector comprises a first DNA segment comprising a 5 ' ITR linked to a second DNA segment comprising a promoter operably linked to a third DNA segment comprising a second open reading frame linked to a fourth DNA segment comprising a 3 'ITR.
  • one rAAV vector comprises a first DNA segment comprising a 5 'ITR linked to a second DNA segment comprising a promoter operably linked to a third DNA segment comprising the 5 ' end of an open reading frame linked to fourth DNA segment comprising a 5 ' splice site linked to a fifth DNA segment comprising a 3 ' ITR.
  • the second rAAV vector comprises a first DNA segment comprising a 5' ITR linked to a second DNA segment comprising a 3' splice site linked to a third DNA segment comprising the 3' end of the open reading frame linked to a fourth DNA segment comprising a 3 'ITR.
  • the host cell is preferably contacted with both of the vectors, concurrently, although it is envisioned that the host cell may be contacted with each vector at a different time relative to the contact with the other vector(s).
  • composition of the invention is administered to the cells or tissues of an animal.
  • rAAV vectors have shown promise in transferring the CFTR gene into airway epithelial cells of animal models and nasal sinus of CF patients.
  • high level expression of CFTR has not been achieved due to the fact that AAV cannot accommodate the full-length CFTR gene together with a potent promoter.
  • a number of studies have tried to optimize rAAV-mediated CFTR expression by utilizing truncated or partially deleted CFTR genes together with stronger promoters.
  • the present invention includes the administration to an animal of a composition of the invention comprising at least two rAAV vectors which together encode CFTR.
  • the present invention is useful to overcome the current size limitation for transgenes within rAAV vectors, and allows for the incorporation of a larger transcriptional regulatory region, e.g., a stronger heterologous promoter or the endogenous CFTR promoter.
  • a larger transcriptional regulatory region e.g., a stronger heterologous promoter or the endogenous CFTR promoter.
  • transgene expression from rAAV luciferase vectors, with or without a promoter can be greatly enhanced by co-infection with an independent rAAV vector carrying the cytomegalovirus (CMV) and simian virus 40 (SV40) enhancers.
  • CMV cytomegalovirus
  • SV40 simian virus 40
  • co-infection with a transgene containing vector and a second vector comprising at least one, preferably at least two or more, enhancer sequences, of cell lines and muscle in vivo resulted in a greater than 600-fold enhancement of transgene expression from a minimal SV40 promoter.
  • 200-fold enhancement was also achieved by cis- activation of ITRs in transgene containing vectors without a promoter.
  • large regulatory elements including tissue specific enhancers can be introduced into cells by a separate rAAV to regulate the expression of a second transgene containing vector in cis following intracellular concatamerization.
  • the invention provides a composition comprising at least two recombinant AAV genomes.
  • the composition comprises a first recombinant AAV comprising a first recombinant DNA molecule comprising linked: i) a first DNA segment comprising a 5 '-inverted terminal repeat of AAV; ii) a second DNA segment which does not comprise AAV sequences; and iii) a third DNA segment comprising a 3 '-inverted terminal repeat of AAV; and comprises a second recombinant AAV comprising a second recombinant DNA molecule comprising linked: i) a first DNA segment comprising a 5 '-inverted terminal repeat of AAV; ii) a second DNA segment which does not comprise AAV sequences and which second DNA segment is different than the second DNA segment of the first recombinant DNA molecule; and iii) a third DNA segment comprising a 3 '-inverted terminal repeat of AAV.
  • composition of the invention is preferably contacted with a mammalian host cell, e.g., a murine, canine, feral or human cell.
  • a host cell may be contacted with each recombinant AAV individually, e.g., sequentially.
  • a host cell is contacted with at least two recombinant AAV genomes.
  • a first recombinant AAV comprises a first recombinant DNA molecule comprising linked ) a first DNA segment comprising a 5 '-inverted terminal repeat of AAV; ii) a second DNA segment which does not comprise AAV sequences; and iii) a third DNA segment comprising a 3 '-inverted terminal repeat of AAV.
  • a second recombinant AAV comprises a second recombinant DNA molecule comprising linked: i) a first DNA segment comprising a 5 '-inverted terminal repeat of AAV; ) a second DNA segment which does not comprise AAV sequences and which second DNA segment is different than the second DNA segment of the first recombinant DNA molecule; and iii) a third DNA segment comprising a 3 '-inverted terminal repeat of AAV.
  • the second DNA segment of the first recombinant DNA molecule comprises a portion of an open reading frame, e.g., an exon of a multi-exon gene, operably linked to a promoter.
  • the promoter may be the endogenous promoter for the gene corresponding to the open reading frame.
  • the second DNA segment of the second recombinant DNA molecule comprises the remainder of the open reading frame which together with the second DNA segment of the first recombinant DNA molecule encodes a full-length polypeptide.
  • the first recombinant DNA molecule comprises a splice donor site 3' to the open reading frame
  • the second DNA segment of the second recombinant DNA molecule comprises a splice acceptor site 5' to the remainder of the open reading frame
  • the second DNA segment of the first recombinant DNA molecule comprises at least one heterologous enhancer and/or at least one heterologous promoter, i.e., the enhancer and/or promoter sequences are not derived from AAV sequences.
  • the second DNA segment of the second recombinant DNA molecule comprises at least a portion of an open reading frame.
  • the second DNA segment of the first recombinant DNA molecule comprises an origin of replication functional in a host cell, e.g., a viral origin of replication such as OriP.
  • the origin is functional in a human cell.
  • the second DNA segment of the first recombinant DNA molecule further comprises DNA encoding a protein that binds to the origin of replication, e.g., EBNA-1.
  • the second DNA segment in the second recombinant DNA molecule comprises at least a portion of an open reading frame, and preferably a promoter operably linked to the open reading frame.
  • the second DNA segment of the first recombinant DNA molecule comprises a cis-act ⁇ ng integration sequence(s) for a recombinase and also encodes a recombinase or integrase that is specific for the integration sequence(s), e.g., Cre/lox system of bacteriophage PI (U.S. Patent No. 5,658,772), the FLP/FRT system of yeast, the Gin recombinase of phage Mu, the Pin recombinase of E. coli, the R/RS system of the pSRl plasmid, a retrotransposase or the integrase from a lentivirus or retro virus.
  • Cre/lox system of bacteriophage PI U.S. Patent No. 5,658,772
  • the FLP/FRT system of yeast the Gin recombinase of phage Mu
  • the Pin recombinase of E. coli the R/RS
  • the second DNA segment in the second recombinant DNA molecule comprises at least a portion of an open reading frame, and preferably a promoter operably linked to the open reading frame.
  • FIG. 1 Structure of proviral shuttle vector and the predicted structure of rAAV circular intermediate monomers. With the aid of a rAAV c/s-acting plasmid, pCisAV.GFP3ori (Panel A), AV.GFP3ori recombinant virus was produced (Panel B). This vector encoded a GFP transgene cassette, an ampicillin resistance gene (amp), and a bacterial replication origin (ori). The predominant form of circular intermediates isolated following transduction of Hela cells with AV.GFP3ori consisted of head- to-tail monomers (Panels C and D).
  • Figure 2 Structural analysis of rAAV circular intermediates in Hela cells.
  • Circular rAAV intermediate clones isolated from AV.GFP3ori infected Hela cells were analyzed by diagnostic restriction digestion with Asel, Sphl, and Pstl together with Southern blotting against ITR, GFP, and Stuffer 32 P-labeled probes.
  • panel A four clones representing the diversity of intermediates found ( i 90, p333, p280, and p345) gave a diagnostic Pstl (P) restriction pattern (3 kb and 1.7 kb bands) consistent with a circular monomer or multimer intact genome [agarose gel (Left) and Southern blot (Right)].
  • Sphl (S) digestion demonstrated existence of a single ITR (pi 90), two ITRs in a head-to-tail orientation (p333 and p280), and three ITRs (p345) in isolated circular intermediates.
  • the restriction pattern of pCisAV.GFP3ori (U; uncut, P; Pstl cut, S; Sphl cut) and 1 kb DNA ladder (L) are also given for comparison.
  • One additional circular form (p340) was repetitively seen and had an unidentifiable structure which lacked intact ITR sequences. Circular concatamers were identified by partial digestion with Asel for clones p280 (dimer) and p333 (monomer) as is shown in Panel B.
  • FIG. 3 Adenovirus augments AAV circular intermediate formation in Hela cells.
  • Infection of Hela cells with increasing doses (0, 500, and 5000 particles/cell) of recombinant El -deleted adenovirus (Ad.CMVlacZ) leads to substantial expression of E2a 72kd DNA Binding Protein, as demonstrated by indirect immunofluorescent staining for DBP at 72 hours post-infection (Panel A).
  • Co-infection of Hela cells with Ad.CMVlacZ (5000 particles/cell) and AV.GFP3ori 1000 DNA particles/cell led to substantial augmentation of rAAV GFP transgene expression (Panel B).
  • Hirt DNAs from AV.GFP3ori 1000 DNA particles/cell infected Hela cells with or without co-infection with Ad.CMVlacZ (5,000 particles/cell) were used to transform E. coli.
  • Panel F depicts the abundance of head-to-tail circular intermediates as a percentage of total ampicillin-resistant bacterial CFU isolated from Hirt DNA.
  • FIG. 4 Formation of rAAV head-to-tail circular intermediates following in vivo transduction of muscle.
  • the tibialis anterior muscle of 4-5 week old C57BL/6 mice were infected with AV.GFP3ori (3 X 1010 particles) in HEPES buffered saline (30 ⁇ l).
  • GFP expression (Panel A) was analyzed by direct immunofluorescence of freshly excised tissues and/or in formalin- fixed cryopreserved tissue sections in four independently injected muscles harvested at 0, 5, 10, 16, 22 and 80 days post-infection. GFP expression was detected at low levels beginning at 10 days and was maximum at 22 days post-infection.
  • Hirt DNA was isolated from muscle samples at each of the various time points and after points was used to transform E. coli.
  • Rescued plasmids (p439, pi 6, pi 7) were analyzed by Southern blotting in Panel B showing an agarose gel on left and ITR probed blot on right. U:uncut, P:PstI cut, and S:SphI cut.
  • the schematic drawing of the most predominant type of head-to-tail circular AAV intermediate plasmids rescued from bacteria is given in the right of Panel B and shows the structure of pi 7 as an example.
  • FIG. 5 Frequency of circular intermediate formation in muscle following transduction with rAAV.
  • Hirt DNAs isolated from rAAV infected tibialis muscle were used to transform E. coli and the rescued plasmids analyzed by Southern blotting (greater than 20 clones were analyzed from at least two independent muscle samples for each time point).
  • the averages of total head-to-tail circular intermediate clones (line) and ampicillin resistant bacterial clones (bar) isolated from each tibialis anterior muscle at 0, 5, 10, 16, 22 and 80 days post-infection are summarized in Panel A. Only plasmids which contained 1-2 ITRs were included in the estimation of total head- to-tail circular intermediates.
  • Panel B demonstrates the diversity of ITR arrays found in head-to-tail circular intermediates at 80 days post-infection. This panel depicts a Southern blot probed with ITR sequences and represents circular intermediates with 1 -3 ITRs. Sphl fragments which hybridize to ITR probes indicate the size of inverted ITR arrays (marked by arrows to right of gel). Additional restriction enzyme analysis was used to determine the structure of monomer and multimer circular intermediates. Examples are shown for two multimer (pi 36 and pi 43) circular intermediates which contain approximately three AAV genomes.
  • Undigested plasmids of pi 36 and pi 43 migrate greater than 12 kb and is contrasted to the most predominant form of head-to-tail undigested circular intermediates at 22 days which migrate at 2.5 kb.
  • the digestion pattern of pi 36 is consistent with a uniform head-to-tail configuration of three genomes which is indistinguishable from digestion patterns of pi 39 which contains one circularized genome (undigested pi 39 migrates at 2.5 kb, data not shown, also see examples pl7 in Figure 4).
  • pi 36 depicts a more complex head-to-tail multimer circular intermediate which has various deletions and duplications within the ITR arrays. Predicted structure of five representative intermediates is schematically shown in Panel C.
  • FIG. 6 Molecular size of circular intermediates in muscle. Hirt DNA from AV.GFP3ori infected muscle was size fractionated by electrophoresis and various molecular weight fractions transformed into E. coli. Results demonstrate the abundance of circular intermediates at each of the given molecular weights at 22 and 80 days post- infection with the rAAV shuttle vector. Structure of circular intermediates were confirmed by Southern blot restriction analysis. Figure 7. Head- to-tail circular intermediates demonstrate increased stability of GFP expression following transient transfection in Hela cells. Subconfluent monolayers of Hela cells were co-transfected with p81, p87, or pCMVGFP and pRSVlacZ as an internal control for transfection efficiency as described in the methods.
  • Panel A demonstrates the expansion of GFP clones after one passage (arrows). Quantification of clone size and numbers are shown in Panel B. Clone size represents the mean raw values while clone numbers are normalized for transfection efficiency as determined by X-gal staining for pRSVlacZ. The data at the top of bar graph values for each construct in Panel B represents quantification of GFP clones after second passage (also normalized for transfection efficiency). Results indicate the mean (+/-SEM) of duplicate experiments with greater than 20 fields quantified for each experimental point.
  • results in Panel D compare the extent of luciferase transgene expression following transfection with pGL3 and pGL3(ITR) at 10 days (passage-2) post-transfection. Results are the mean (+/-SEM) for triplicate experiments and are normalized for transfection efficiency using a dual renilla luciferase reporter vector (pRLSV40, Promega).
  • FIG. 1 Identification of adenoviral genes responsible for augmentation of AAV circular intermediate formation. Hela cells were infected with
  • AV.GFP3ori 1000 DNA particles/cell in the presence of wtAd5, /802 (E2a- deleted), and d ⁇ 1004 (E4-deleted) adenovirus (at the indicated MOIs).
  • Panel B depicts results from Southern blot analysis of Hirt DNA following hybridization to a GFP P 32 -labeled probe. DNA loads were 10%) of the total Hirt yield from a 35 mm plate of Hela cells.
  • FIG. 9 Model for independent mechanistic interactions of adenovirus with lytic and latent phase aspects of the AAV life cycle.
  • the adenoviral E4 gene has been shown to augment the level of rAAV second strand synthesis giving rise to replication form dimers (Rf d ) and monomers (Rf m ) ( Figure 8B). This augmentation leads to substantial increases in transgene expression from rAAV vectors and most closely mirrors lytic phase replication of wtAAV as head-to-head and tail-to-tail concatamers. In contrast, E4 expression inhibits the formation of head-to-tail circular intermediates of AAV.
  • these circular intermediates may represent pre-integration complexes as previously hypothesized for Rep facilitated integration.
  • circular intermediates may accumulate episomally in rAAV infected cells.
  • FIG. 10 Individual chemical sequence of Sphl fragments from p81 (A; SEQ ID NO:4), p79 (B; SEQ ID NO:5), and pl202 (C; SEQ ID NO:6) AAV circular intermediates.
  • the ends of the sequence represent Sphl restriction enzyme sites within head-to-tail circular AAV genomes cloned with the AV-GFP3ori shuttle virus.
  • Figure 1 Chemical sequence homology of three AAV circular intermediates with various conformations of ITR arrays (SEQ ID NO:4, SEQ ID NO: 5 and SEQ ID NO: 6). Diversity in ITR arrays are evident from the non- conserved bases marked in lower case. The ends of the sequence (underlined) represent Sphl restriction enzyme sites within head-to-tail circular AAV genomes cloned with the AV.GFP3ori shuttle virus.
  • FIG. 12 Palindromic repeat structure derived from chemical sequencing of AAV circular intermediate isolate p81. Secondary structure of the sense strand is depicted in the top box with plasmid reference given below.
  • Figure 12B Palindromic repeat structure derived from chemical sequencing of AAV circular intermediate isolate p79. Secondary structure of the sense strand is depicted in the top box with plasmid reference given below.
  • Figure 12C Palindromic repeat structure derived from chemical sequencing of AAV circular intermediate isolate p79. Secondary structure of the sense strand is depicted in the top box with plasmid reference given below.
  • Figure 13 Persistence of GFP expression in developing Xenopus embryos microinjected with AAV circular intermediate isolate p81. The extent of GFP fluorescence in tadpoles reflects the stability of episomal or integrated microinjected plasmids. Bright field image on the left is of the p81 injected embryo.
  • the p81 injected embryo depicts fluorescence in nearly all cells by one week post-injection. In contrast, a mosaic pattern of expression in a minority of cells in pCisAV.GFPori injected embryos.
  • the pCisAV.GFPori plasmid contains the identical promoter sequences driving GFP gene expression and two ITRs separated by stuffer sequence.
  • Figure 14 Mechanistic scheme for determining pathways for rAAV circular concatamer formation.
  • the two independent vectors used in these studies, AV. Alkphos and AV.GFP3.ori, are shown in Panel A . Restriction sites important in the structural analysis of circular intermediates are also shown.
  • Panel B a schematic representation of two potential models for circular concatamer formation is depicted, along with the methods to experimentally differentiate which of these processes is active in muscle. Following co- infection of the tibialis muscle with AV. Alkphos and AV.GFP3.ori, all subsequently rescued plasmids arise solely from circular intermediates containing AV.GFP3ori genomes.
  • FIG. 15 Co-infection of tibialis muscle of mice with AV. Alkphos and AV.GFP3ori.
  • Transgene expression of rAAV infected tibialis muscle was determined at 14, 35, 80 (Panels A and A'), and 120 (Panels B-D) days following co-infection with 5 x 10 9 DNA particles each of AV. Alkphos and AV.GFP3ori.
  • the time course of transgene expression started around 14 days and peaked by 35-80 days.
  • the extent of co-infection of myo fibers with both Alkphos and GFP rAAV was determined in serial sections of 80 and 120 day post-infection muscle samples.
  • Panels A-C represent GFP fluorescence of formalin fixed, cryoprotected sections, while panels A'-C depict the histochemical staining for Alkaline phosphatase in adjacent serial sections.
  • a short staining time (7 minutes) was necessary to observe variation in staining levels for comparison to GFP. It was found that longer staining times (30 minutes) saturated the Alkphos signal.
  • the boxed region in panels B and B' are enlarged in panels C and C, respectively.
  • a more precise correlation of GFP and Alkphos staining in myo fibers is given in Panel D in which co-localization of GFP and Alkphos expression was examined in the same section of a 120 day post-infected sample.
  • the left panel of D shows a high power Nomarski photomicrograph of a group of myo fibers (traced in red) , while the corresponding GFP and Alkphos staining patterns are shown in the right panel.
  • Photomicrographs of Alkphos staining were taken with a red filter to allow for superimposition of staining patterns with GFP fluorescence.
  • Co-expression of Alkphos and GFP is shown within myo fibers as a yellow/orange color.
  • Myo fibers are marked as follows: (-) negative for both Alkphos and GFP, (*) positive for only GFP, and (+) positive for both GFP and Alkphos.
  • Figure 16 Rescue of circular intermediates and characterization of DNA hybridization patterns.
  • amp ampicillin resistance gene
  • bacterial ori inco ⁇ orated into the AV.GFP3ori vector
  • the extent of circular intermediate formation was assessed by rescuing amp resistant plasmids following transformation of 1/5 the isolated Hirt DNA into E. coli Sure cells.
  • Twenty plasmids from each muscle sample were prepared and analyzed by slot blot hybridization against GFP, Alkphos, and Amp 32 P-labeled DNA probes. A representative group demonstrating the hybridization patterns is shown in Panel A.
  • Panel B depicts the mean (+/-SEM) number of rescued bacterial plasmids that hybridized to either GPF alone, or to both GFP and Alkphos probes, following transformation of l/5 th of the Hirt DNA. These numbers were calculated from the percentage of plasmids hybridizing to GPF and/or Alkphos and the total CFU plating efficiency derived from the original transformation. In total, 3 independent muscle samples were analyzed for a total of 60 plasmids at each time point. The percentage of GFP hybridization positive rescued plasmids that also demonstrated hybridization to Alkphos is shown in Panel C. These data demonstrate an increase in the abundance of rescued GFP/ Alkphos co-encoding circular intermediates over time.
  • FIG. 17 Transgene expression from rescued circular intermediates.
  • Rescued circular intermediate plasmids were transfected into 293 cells for assessment of their ability to express encoded transgenes.
  • all GFP hybridization positive clones from at least two muscles were tested for each time point and scored for their ability to express GFP and Alkaline phosphatase. In total at least 40 clones were evaluated for each time point.
  • Three patterns of transgene expression were observed following transfection of these plasmids: I) no gene expression (Panel A), II) GFP expression only (Panel B), and III) GFP and Alkphos expression (Panel C).
  • Panels A-C depict Nomarski photomicrographs (left) of GFP fluorescent fields (center) and Alkphos staining of a different field from the same culture (right). The percentage of GFP hybridization positive clones that also expressed GFP is shown in Panel D.
  • this panel illustrates the percentage of GFP expressing clones also expressing Alkphos.
  • FIG. 18 Structural analysis of bi-functional concatamer circular intermediates. To fully characterize the nature of GFP and Alkphos co- expressing circular intermediates, detailed structural analyses were performed using restriction enzyme mapping and Southern blot hybridization with GFP, Alkphos, and ITR 32 P-labeled probes. Results from Southern blot analysis of plasmid clone #33 (Panel A) and clone #5 (Panel C) are given as representative examples of circular intermediates isolated from 80 and 35 day Hirt DNA of rAAV infected muscle, respectively. Agarose gels were run in triplicate for each of these clones and Southern blot filters were hybridized with one of the three DNA probes as indicated below each autoradiogram.
  • Molecular weights are indicated to the left of the ethidium stained agarose gel and restriction enzymes are marked on the top of each gel/filter.
  • Panels B and D give the deduced structure of plasmid clones #33 and #5, respectively, as based on Southern blot analysis.
  • the position of restriction enzyme sites are marked with the indicated orientation of intact viral genomes.
  • ITR arrays may be in a double-D structure (i.e., one ITR flanked by two D- sequence rather than two ITRs) and hence ITR arrays containing fragments may appear 147 bp shorter than indicated.
  • ITR arrays containing fragments may appear 147 bp shorter than indicated.
  • the position of 5 'and 3' ITRs is indicated rather than representing a single ITR at these junctions.
  • FIG. 19 Application of rAAV circular concatamers to deliver trans- splicing vectors with large gene inserts.
  • Panel A depicts two rAAV vectors encoding two halves of a cDNA (red) and flanked by splice site consensus sequences (brown).
  • Panel B depicts one potential type of intermolecular concatamer following co-infection of cells with the independent vectors shown in panel A. Full length transgene mRNA can then be produced by splicing.
  • Panel C depicts two rAAV vectors encoding two halves of a CFTR DNA flanked by a promoter and splice donor or a splice acceptor and a poly A sequence, respectively.
  • Panel D shows one potential type of intermolecular concatamer following co-infection of cells with the vectors in Panel C.
  • Figure 20 Schematic representation of the rAAV vectors used for cis- activation.
  • Figure 21 Strategy for enhancing rAAV gene expression through intermolecular c/s-activation.
  • Two independent rAAV viruses one encoding a transgene with or without a minimal promoter (e.g., AV.SV(P)Luc) and another harboring enhancer sequences (e.g., AV.SupEnh), were used to co-infect the same tissue.
  • Subsequent concatamerization between two rAAV vectors substantially augments expression of the transgene, due to the presence of enhancer elements within the same circularized molecule.
  • Figure 22 Intermolecular c/s-activation increases rAAV mediated gene transfer in fibroblasts.
  • Human fibroblast cells were infected with the indicated rAAV vector(s) at an moi of 1000 for each individual vector. Luciferase activity was examined at 3 days post-infection. The data represent the mean +/- SEM of 6 independent samples for each experimental condition.
  • FIG. 23 Intermolecular cis-activation increases rAAV mediated gene transfer to muscle in vivo.
  • Mouse tibialis anterior muscles were infected with the indicated rAAV vector(s) at 2 x 10 10 particles per viral vector in a total volume of 30 ⁇ l PBS.
  • the luciferase activity in rAAV infected or mock infected (PBS) muscles was examined at 30 days (Panel A) and 90 days (Panel B) post- infection.
  • the data represent the mean +/- SEM of 6 independent muscle samples for each experimental condition.
  • Co-administration of the AV.SupEnh vector harboring enhancer elements substantially enhanced rAAV mediated luciferase expression in muscle from both the ITR and the minimal SV40 promoter.
  • FIG. 24 Viral constructs for the generation of autonomously replicating rAAV vectors as circular concatamers.
  • Panel A depicts two rAAV constructs used to test this hypothesis. One encodes the GFP transgene (green) and the other encodes the EBNA-1 (red) and OriP (purple) sequences necessary for autonomous replication. Additionally, sequences encoded within the GFP vector allow for rescue of circular intermediates in bacteria.
  • Panel B depicts one potential type of intermolecular concatamer following co-infection of the independent vectors shown in panel A.
  • FIG. 25 rAAV vectors used to generate a trans-splicing vector expressing genomic epo DNA.
  • Panel A shows a schematic of the vectors. An IRES sequence and EGFP gene are included in one of the vectors to allow for direct visualization of transgene expression.
  • Panel B depicts a potential circular concatamer formed after co-infection. The dashed lines indicate the splicing pattern.
  • Panel C shows the hnRNA, splicing pattern and mature mRNA transcripts which result from circular concatamerization of the two vectors.
  • FIG 26 Production of full length Epo protein following co-infection of primary fibroblasts with two independent trans-splicing vectors.
  • Confluent primary fibroblasts (8 x 10 5 cells) were infected with 7 x 10 9 particles of each AV.Epol and/or AV.Epo2.
  • Figure 27 Functional expression of human Epo in vivo using trans- splicing AAV vectors.
  • isolated and/or purified refer to in vitro preparation, isolation and/or purification of a nucleic acid molecule of the invention, so that it is not associated with in vivo substances.
  • a DNA molecule, sequence or segment of the invention preferably is biologically active.
  • a biologically active DNA molecule of the invention has at least about 1%, more preferably at least about 10%, and more preferably at least about 50%>, of the activity of a DNA molecule comprising ITR sequences from a circular intermediate of AAV, e.g., a DNA molecule comprising SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, or a subunit or variant thereof.
  • the activity of a nucleic acid molecule of the invention can be measured by methods well known to the art, some of which are described hereinbelow.
  • the presence of the DNA molecule in a recombinant nucleic acid molecule in a host cell results in episomal persistence and/or increased abundance of the recombinant molecule in those cells relative to corresponding cells having a recombinant nucleic acid molecule lacking a DNA molecule of the invention.
  • a variant DNA molecule, sequence or segment of the invention has at least about 70%, preferably at least about 80%>, and more preferably at least about 90%, but less than 100%, contiguous nucleotide sequence homology or identity to a DNA molecule comprising ITR sequences from a circular intermediate of AAV, e.g., SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, a subunit thereof.
  • a variant DNA molecule of the invention may include nucleotide bases not present in SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, e.g., 5', 3' or internal deletions or insertions, such as the insertion of a restriction endonuclease recognition site, so long as these bases do not substantially reduce the biological activity of the molecule.
  • a substantial reduction in activity means a reduction in activity of greater than about 50%, preferably greater than about 90%.
  • Sources of the Nucleic Acid Molecules of the Invention include AAV infected cells, e.g., any vertebrate, preferably mammalian, cellular source.
  • isolated and/or purified refer to in vitro isolation of a nucleic acid, e.g., DNA molecule from its natural cellular environment, and from association with other components of the cell, such as nucleic acid or polypeptide, so that it can be sequenced, replicated, and/or expressed.
  • a nucleic acid e.g., DNA molecule from its natural cellular environment
  • other components of the cell such as nucleic acid or polypeptide
  • isolated nucleic acid is RNA or DNA containing greater than about 50, preferably about 300, and more preferably about 500 or more, sequential nucleotide bases that comprise a DNA segment from a circular intermediate of AAV which contains at least a portion of the 5' and 3' ITRs and the D sequence, or a variant thereof, that is complementary or hybridizes, respectively, to AAV ITR DNA and remains stably bound under stringent conditions, as defined by methods well known in the art, e.g., in Sambrook et al., 1989.
  • the RNA or DNA is "isolated” in that it is free from at least one contaminating nucleic acid with which it is normally associated in the natural source of the RNA or DNA and is preferably substantially free of any other mammalian RNA or DNA.
  • the phrase "free from at least one contaminating source nucleic acid with which it is normally associated” includes the case where the nucleic acid is reintroduced into the source or natural cell but is in a different chromosomal location or is otherwise flanked by nucleic acid sequences not normally found in the source cell, e.g., in a vector or plasmid.
  • nucleic acid within the scope of the invention is nucleic acid that shares at least about 80%, preferably at least about 90%, and more preferably at least about 95%, sequence identity with SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6, or a subunit thereof.
  • recombinant nucleic acid or "preselected nucleic acid,” e.g., “recombinant DNA sequence or segment” or “preselected DNA sequence or segment” refers to a nucleic acid, e.g., to DNA, that has been derived or isolated from any appropriate cellular source, that may be subsequently chemically altered in vitro, so that its sequence is not naturally occurring, or corresponds to naturally occurring sequences that are not positioned as they would be positioned in a genome which has not been transformed with exogenous DNA.
  • An example of preselected DNA "derived” from a source would be a DNA sequence that is identified as a useful fragment within a given organism, and which is then chemically synthesized in essentially pure form.
  • DNA "isolated" from a source would be a useful DNA sequence that is excised or removed from said source by chemical means, e.g., by the use of restriction endonucleases, so that it can be further manipulated, e.g., amplified, for use in the invention, by the methodology of genetic engineering.
  • recovery or isolation of a given fragment of DNA from a restriction digest can employ separation of the digest on polyacrylamide or agarose gel by electrophoresis, identification of the fragment of interest by comparison of its mobility versus that of marker DNA fragments of known molecular weight, removal of the gel section containing the desired fragment, and separation of the gel from DNA.
  • Preselected DNA includes completely synthetic DNA sequences, semi-synthetic DNA sequences, DNA sequences isolated from biological sources, and DNA sequences derived from RNA, as well as mixtures thereof.
  • Nucleic acid molecules having base pair substitutions are prepared by a variety of methods known in the art. These methods include, but are not limited to, isolation from a natural source (in the case of naturally occurring sequence variants) or preparation by oligonucleotide-mediated (or site- directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of the nucleic acid molecule.
  • Oligonucleotide-mediated mutagenesis is a preferred method for preparing substitution variants. This technique is well known in the art as . described by Adelman et al., DNA, 2, 183 (1983). Briefly, AAV DNA is altered by hybridizing an oligonucleotide encoding the desired mutation to a DNA template, where the template is the single-stranded form of a plasmid or bacteriophage containing the unaltered or native DNA sequence of AAV. After hybridization, a DNA polymerase is used to synthesize an entire second complementary strand of the template that will thus inco ⁇ orate the oligonucleotide primer, and will code for the selected alteration in the AAV DNA.
  • oligonucleotides of at least 25 nucleotides in length are used.
  • An optimal oligonucleotide will have 12 to 15 nucleotides that are completely complementary to the template on either side of the nucleotide(s) coding for the mutation. This ensures that the oligonucleotide will hybridize properly to the single-stranded DNA template molecule.
  • the oligonucleotides are readily synthesized using techniques known in the art such as that described by Crea et al., Proc. Natl. Acad. Sci. U.S. A.. 15., 5765 (1978).
  • the DNA template can be generated by those vectors that are either derived from bacteriophage M13 vectors (the commercially available M13mpl8 and M13mpl9 vectors are suitable), or those vectors that contain a single- stranded phage origin of replication as described by Viera et al., Meth. Enzymol., 151, 3 (1987). Thus, the DNA that is to be mutated may be inserted into one of these vectors to generate single-stranded template. Production of the single- stranded template is described in Sections 4.21-4.41 of Sambrook et al.,
  • single-stranded DNA template may be generated by denaturing double-stranded plasmid (or other) DNA using standard techniques.
  • the oligonucleotide is hybridized to the single- stranded template under suitable hybridization conditions.
  • a DNA polymerizing enzyme usually the Klenow fragment of DNA polymerase I, is then added to synthesize the complementary strand of the template using the oligonucleotide as a primer for synthesis.
  • a heteroduplex molecule is thus formed such that one strand of DNA encodes the mutated form of AAV, and the other strand (the original template) encodes the native, unaltered sequence of AAV.
  • This heteroduplex molecule is then transformed into a suitable host cell, usually a prokaryote such as E. coli JM101.
  • the cells are grown, they are plated onto agarose plates and screened using the oligonucleotide primer radiolabeled with 32-phosphate to identify the bacterial colonies that contain the mutated DNA.
  • the mutated region is then removed and placed in an appropriate vector, generally an expression vector of the type typically employed for transformation of an appropriate host.
  • the method described immediately above may be modified such that a homoduplex molecule is created wherein both strands of the plasmid contain the mutations(s).
  • the modifications are as follows:
  • the single-stranded oligonucleotide is annealed to the single-stranded template as described above.
  • a mixture of three deoxyribonucleotides, deoxyriboadenosine (dATP), deoxyriboguanosine (dGTP), and deoxyribothymidine (dTTP) is combined with a modified thiodeoxyribocytosine called dCTP-( S) (which can be obtained from the Amersham Co ⁇ oration). This mixture is added to the template- oligonucleotide complex.
  • this new strand of DNA Upon addition of DNA polymerase to this mixture, a strand of DNA identical to the template except for the mutated bases is generated.
  • this new strand of DNA will contain dCTP-( ⁇ S) instead of dCTP, which serves to protect it from restriction endonuclease digestion.
  • the template strand can be digested with ExoIII nuclease or another appropriate nuclease past the region that contains the site(s) to be mutagenized. The reaction is then stopped to leave a molecule that is only partially single-stranded.
  • a complete double-stranded DNA homoduplex is then formed using DNA polymerase in the presence of all four deoxyribonucleotide triphosphates, ATP, and DNA ligase.
  • This homoduplex molecule can then be transformed into a suitable host cell such as E. coli JM101.
  • a preferred embodiment of the invention is an isolated and purified DNA molecule comprising a DNA segment comprising SEQ ID NO: 3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, a subunit thereof or a variant thereof having nucleotide substitutions, or deletions or insertions.
  • TT Preparation of Molecules Useful to Practice the Methods of the Invention A. Nucleic Acid Molecules
  • the recombinant or preselected DNA sequence or segment may be circular or linear, double-stranded or single-stranded.
  • the preselected DNA sequence or segment is in the form of chimeric DNA, such as plasmid DNA, that can also contain coding regions flanked by control sequences which promote the expression of the preselected DNA present in the resultant cell line.
  • chimeric means that a vector comprises DNA from at least two different species, or comprises DNA from the same species, which is linked or associated in a manner which does not occur in the "native" or wild type of the species.
  • the preselected DNA may serve a regulatory or a structural function.
  • the preselected DNA may itself comprise a promoter that is active in mammalian cells, or may utilize a promoter already present in the genome that is the transformation target.
  • promoters include the CMV promoter, as well as the SV40 late promoter and retroviral LTRs (long terminal repeat elements), although many other promoter elements well known to the art may be employed in the practice of the invention.
  • Other elements functional in the host cells such as introns, enhancers, polyadenylation sequences and the like, may also be a part of the preselected DNA. Such elements may or may not be necessary for the function of the DNA, but may provide improved expression of the DNA by affecting transcription, stability of the mRNA, or the like. Such elements may be included in the DNA as desired to obtain the optimal performance of the transforming DNA in the cell.
  • Control sequences is defined to mean DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism.
  • the control sequences that are suitable for prokaryotic cells include a promoter, and optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • "Operably linked” is defined to mean that the nucleic acids are placed in a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a peptide or polypeptide if it is expressed as a preprotein that participates in the secretion of the peptide or polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • "operably linked” means that the DNA sequences being linked are contiguous and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accord with conventional practice.
  • the preselected DNA to be introduced into the cells further will generally contain either a selectable marker gene or a reporter gene or both to facilitate identification and selection of transformed cells from the population of cells sought to be transformed.
  • the selectable marker may be carried on a separate piece of DNA and used in a co-transformation procedure.
  • Both selectable markers and reporter genes may be flanked with appropriate regulatory sequences to enable expression in the host cells.
  • Useful selectable markers are well known in the art and include, for example, antibiotic and herbicide-resistance genes, such as neo, hpt, dhfr, bar, aroA, dapA and the like. See also, the genes listed on Table 1 of Lundquist et al. (U.S. Patent No.
  • Reporter genes are used for identifying potentially transformed cells and for evaluating the functionality of regulatory sequences. Reporter genes which encode for easily assayable proteins are well known in the art. In general, a reporter gene is a gene which is not present in or expressed by the recipient organism or tissue and which encodes a protein whose expression is manifested by some easily detectable property, e.g., enzymatic activity. Preferred genes include the chloramphenicol acetyl transferase gene (cat) from Tn9 of E. coli, the beta-glucuronidase gene (gus) of the uidA locus of E. coli, and the luciferase gene from firefly Photinus pyralis.
  • Expression of the reporter gene is assayed at a suitable time after the DNA has been introduced into the recipient cells.
  • the general methods for constructing recombinant DNA which can transform target cells are well known to those skilled in the art, and the same compositions and methods of construction may be utilized to produce the DNA useful herein. For example, J. Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press (2d ed., 1989), provides suitable methods of construction. 2. Transformation into Host Cells
  • the recombinant DNA can be readily introduced into the host cells, e.g., mammalian, bacterial, yeast or insect cells by transfection with an expression vector of the invention, by any procedure useful for the introduction into a particular cell, e.g., physical or biological methods, to yield a transformed cell having the recombinant DNA stably integrated into its genome or present as an episome which can persist in the transformed cells, so that the DNA molecules, sequences, or segments, of the present invention are maintained and/or expressed by the host cell.
  • the host cells e.g., mammalian, bacterial, yeast or insect cells by transfection with an expression vector of the invention, by any procedure useful for the introduction into a particular cell, e.g., physical or biological methods, to yield a transformed cell having the recombinant DNA stably integrated into its genome or present as an episome which can persist in the transformed cells, so that the DNA molecules, sequences, or segments, of the present invention are maintained and/or expressed by the host cell.
  • Physical methods to introduce a preselected DNA into a host cell include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like.
  • Biological methods to introduce the DNA of interest into a host cell include the use of DNA and RNA viral vectors.
  • the main advantage of physical methods is that they are not associated with pathological or oncogenic processes of viruses. However, they are less precise, often resulting in multiple copy insertions, random integration, disruption of foreign and endogenous gene sequences, and unpredictable expression.
  • the term "cell line” or "host cell” is intended to refer to well-characterized homogenous, biologically pure populations of cells. These cells may be eukaryotic cells that are neoplastic or which have been “immortalized” in vitro by methods known in the art, as well as primary cells, or prokaryotic cells.
  • the cell line or host cell is preferably of mammalian origin, but cell lines or host cells of non-mammalian origin may be employed, including plant, insect, yeast, fungal or bacterial sources.
  • the preselected DNA sequence is related to a DNA sequence which is resident in the genome of the host cell but is not expressed, or not highly expressed, or, alternatively, overexpressed.
  • Transfected or “transformed” is used herein to include any host cell or cell line, the genome of which has been altered or augmented by the presence of at least one preselected DNA sequence, which DNA is also referred to in the art of genetic engineering as “heterologous DNA,” “recombinant DNA,”
  • exogenous DNA "genetically engineered,” “non-native,” or “foreign DNA,” wherein said DNA was isolated and introduced into the genome of the host cell or cell line by the process of genetic engineering.
  • the host cells of the present invention are typically produced by transfection with a DNA sequence in a plasmid expression vector, a viral expression vector, or as an isolated linear DNA sequence.
  • RNA produced from introduced DNA segments include, for example, “molecular biological” assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR; “biochemical” assays, such as detecting the presence of a polypeptide expressed from a gene present in the vector, e.g., by immunological means (immunoprecipitations, imrnunoaffinity columns, ELIS As and Western blots) or by any other assay useful to identify molecules falling within the scope of the invention.
  • immunological means immunological means
  • imrnunoaffinity columns include ELIS As and Western blots
  • RT-PCR may be employed.
  • PCR it is first necessary to reverse transcribe RNA into DNA, using enzymes such as reverse transcriptase, and then through the use of conventional PCR techniques amplify the DNA.
  • PCR techniques while useful, will not demonstrate integrity of the RNA product.
  • Further information about the nature of the RNA product may be obtained by Northern blotting. This technique demonstrates the presence of an RNA species and gives information about the integrity of that RNA. The presence or absence of an RNA species can also be determined using dot or slot blot Northern hybridizations. These techniques are modifications of Northern blotting and only demonstrate the presence or absence of an RNA species.
  • While Southern blotting and PCR may be used to detect the DNA segment in question, they do not provide information as to whether the DNA segment is being expressed. Expression may be evaluated by specifically identifying the polypeptide products of the introduced DNA sequences or evaluating the phenotypic changes brought about by the expression of the introduced DNA segment in the host cell. TTT. Dosages, Formulations and Routes of Administration Administration of a nucleic acid molecule may be accomplished through the introduction of cells transformed with the nucleic acid molecule (see, for example, WO 93/02556), the administration of the nucleic acid molecule itself (see, for example, Feigner et al., U.S. Patent No.
  • Administration of the therapeutic agents in accordance with the present invention may be continuous or intermittent, depending, for example, upon the recipient's physiological condition, whether the pu ⁇ ose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners.
  • the administration of the agents of the invention may be essentially continuous over a preselected period of time or may be in a series of spaced doses. Both local and systemic administration is contemplated.
  • agents of the invention are amenable to chronic use, preferably by systemic administration.
  • One or more suitable unit dosage forms comprising the therapeutic agents of the invention can be administered by a variety of routes including oral, or parenteral, including by rectal, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, intrathoracic, intrapulmonary and intranasal routes.
  • the formulations may, where appropriate, be conveniently presented in discrete unit dosage forms and may be prepared by any of the methods well known to pharmacy. Such methods may include the step of bringing into association the therapeutic agent with liquid carriers, solid matrices, semi-solid carriers, finely divided solid carriers or combinations thereof, and then, if necessary, introducing or shaping the product into the desired delivery system.
  • the therapeutic agents of the invention are prepared for oral administration, they are preferably combined with a pharmaceutically acceptable carrier, diluent or excipient to form a pharmaceutical formulation, or unit dosage form.
  • a pharmaceutically acceptable carrier diluent or excipient to form a pharmaceutical formulation, or unit dosage form.
  • the total active ingredients in such formulations comprise from 0.1 to 99.9%) by weight of the formulation.
  • pharmaceutically acceptable it is meant the carrier, diluent, excipient, and/or salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
  • the active ingredient for oral administration may be present as a powder or as granules; as a solution, a suspension or an emulsion; or in achievable base such as a synthetic resin for ingestion of the active ingredients from a chewing gum.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • compositions containing the therapeutic agents of the invention can be prepared by procedures known in the art using well known and readily available ingredients.
  • the agent can be formulated with common excipients, diluents, or carriers, and formed into tablets, capsules, suspensions, powders, and the like.
  • excipients, diluents, and carriers that are suitable for such formulations include the following fillers and extenders such as starch, sugars, mannitol, and silicic derivatives; binding agents such as carboxymethyl cellulose, HPMC and other cellulose derivatives, alginates, gelatin, and polyvinyl-pyrrolidone; moisturizing agents such as glycerol; disintegrating agents such as calcium carbonate and sodium bicarbonate; agents for retarding dissolution such as paraffin; reso ⁇ tion accelerators such as quaternary ammonium compounds; surface active agents such as cetyl alcohol, glycerol monostearate; adso ⁇ tive carriers such as kaolin and bentonite; and lubricants such as talc, calcium and magnesium stearate, and solid polyethyl glycols.
  • fillers and extenders such as starch, sugars, mannitol, and silicic derivatives
  • binding agents such as carboxymethyl cellulose, HPMC and other
  • tablets or caplets containing the agents of the invention can include buffering agents such as calcium carbonate, magnesium oxide and magnesium carbonate.
  • Caplets and tablets can also include inactive ingredients such as cellulose, pregelatinized starch, silicon dioxide, hydroxy propyl methyl cellulose, magnesium stearate, microcrystalline cellulose, starch, talc, titanium dioxide, benzoic acid, citric acid, corn starch, mineral oil, polypropylene glycol, sodium phosphate, and zinc stearate, and the like.
  • Hard or soft gelatin capsules containing an agent of the invention can contain inactive ingredients such as gelatin, microcrystalline cellulose, sodium lauryl sulfate, starch, talc, and titanium dioxide, and the like, as well as liquid vehicles such as polyethylene glycols (PEGs) and vegetable oil.
  • enteric coated caplets or tablets of an agent of the invention are designed to resist disintegration in the stomach and dissolve in the more neutral to alkaline environment of the duodenum.
  • the therapeutic agents of the invention can also be formulated as elixirs or solutions for convenient oral administration or as solutions appropriate for parenteral administration, for instance by intramuscular, subcutaneous or intravenous routes.
  • the pharmaceutical formulations of the therapeutic agents of the invention can also take the form of an aqueous or anhydrous solution or dispersion, or alternatively the form of an emulsion or suspension.
  • the therapeutic agent may be formulated for parenteral administration (e.g., by injection, for example, bolus injection or continuous infusion) and may be presented in unit dose form in ampules, pre-filled syringes, small volume infusion containers or in multi-dose containers with an added preservative.
  • the active ingredients may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredients may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
  • formulations can contain pharmaceutically acceptable vehicles and adjuvants which are well known in the prior art. It is possible, for example, to prepare solutions using one or more organic solvent(s) that is/are acceptable from the physiological standpoint, chosen, in addition to water, from solvents such as acetone, ethanol, isopropyl alcohol, glycol ethers such as the products sold under the name "Dowanol”, polyglycols and polyethylene glycols, C r C 4 alkyl esters of short-chain acids, preferably ethyl or isopropyl lactate, fatty acid triglycerides such as the products marketed under the name "Miglyol", isopropyl myristate, animal, mineral and vegetable oils and polysiloxanes.
  • organic solvent(s) that is/are acceptable from the physiological standpoint, chosen, in addition to water, from solvents such as acetone, ethanol, isopropyl alcohol, glycol ethers such as the products sold under the name "Dowanol”, polyg
  • compositions according to the invention can also contain thickening agents such as cellulose and/or cellulose derivatives. They can also contain gums such as xanthan, guar or carbo gum or gum arabic, or alternatively polyethylene glycols, bentones and montmorillonites, and the like. It is possible to add, if necessary, an adjuvant chosen from antioxidants, surfactants, other preservatives, film-forming, keratolytic or comedolytic agents, perfumes and colorings. Also, other active ingredients may be added, whether for the conditions described or some other condition.
  • thickening agents such as cellulose and/or cellulose derivatives. They can also contain gums such as xanthan, guar or carbo gum or gum arabic, or alternatively polyethylene glycols, bentones and montmorillonites, and the like. It is possible to add, if necessary, an adjuvant chosen from antioxidants, surfactants, other preservatives, film-forming, keratolytic or comedolytic agents, perfumes
  • the galenical forms chiefly conditioned for topical application take the form of creams, milks, gels, dispersion or microemulsions, lotions thickened to a greater or lesser extent, impregnated pads, ointments or sticks, or alternatively the form of aerosol formulations in spray or foam form or alternatively in the form of a cake of soap.
  • the agents are well suited to formulation as sustained release dosage forms and the like.
  • the formulations can be so constituted that they release the active ingredient only or preferably in a particular part of the intestinal or respiratory tract, possibly over a period of time.
  • the coatings, envelopes, and protective matrices may be made, for example, from polymeric substances, such as polylactide-glycolates, liposomes, microemulsions, microparticles, nanoparticles, or waxes. These coatings, envelopes, and protective matrices are useful to coat indwelling devices, e.g., stents, catheters, peritoneal dialysis tubing, and the like.
  • the therapeutic agents of the invention can be delivered via patches for transdermal administration. See U.S. Patent No. 5,560,922 for examples of patches suitable for transdermal delivery of a therapeutic agent.
  • Patches for transdermal delivery can comprise a backing layer and a polymer matrix which has dispersed or dissolved therein a therapeutic agent, along with one or more skin permeation enhancers.
  • the backing layer can be made of any suitable material which is impermeable to the therapeutic agent.
  • the backing layer serves as a protective cover for the matrix layer and provides also a support function.
  • the backing can be formed so that it is essentially the same size layer as the polymer matrix or it can be of larger dimension so that it can extend beyond the side of the polymer matrix or overlay the side or sides of the polymer matrix and then can extend outwardly in a manner that the surface of the extension of the backing layer can be the base for an adhesive means.
  • the polymer matrix can contain, or be formulated of, an adhesive polymer, such as polyacrylate or acrylate/vinyl acetate copolymer.
  • an adhesive polymer such as polyacrylate or acrylate/vinyl acetate copolymer.
  • Examples of materials suitable for making the backing layer are films of high and low density polyethylene, polypropylene, polyurethane, polyvinylchloride, polyesters such as poly(ethylene phthalate), metal foils, metal foil laminates of such suitable polymer films, and the like.
  • the materials used for the backing layer are laminates of such polymer films with a metal foil such as aluminum foil. In such laminates, a polymer film of the laminate will usually be in contact with the adhesive polymer matrix.
  • the backing layer can be any appropriate thickness which will provide the desired protective and support functions.
  • a suitable thickness will be from about 10 to about 200 microns.
  • those polymers used to form the biologically acceptable adhesive polymer layer are those capable of forming shaped bodies, thin walls or coatings through which therapeutic agents can pass at a controlled rate.
  • Suitable polymers are biologically and pharmaceutically compatible, nonallergenic and insoluble in and compatible with body fluids or tissues with which the device is contacted. The use of soluble polymers is to be avoided since dissolution or erosion of the matrix by skin moisture would affect the release rate of the therapeutic agents as well as the capability of the dosage unit to remain in place for convenience of removal.
  • Exemplary materials for fabricating the adhesive polymer layer include polyethylene, polypropylene, polyurethane, ethylene/propylene copolymers, ethylene/ethylacrylate copolymers, ethylene/vinyl acetate copolymers, silicone elastomers, especially the medical-grade polydimethylsiloxanes, neoprene rubber, polyisobutylene, polyacrylates, chlorinated polyethylene, polyvinyl f chloride, vinyl chloride-vinyl acetate copolymer, crosslinked polymethacrylate polymers (hydrogel), polyvinylidene chloride, poly(ethylene terephthalate), butyl rubber, epichlorohydrin rubbers, ethylenvinyl alcohol copolymers, ethylene- vinyloxy ethanol copolymers; silicone copolymers, for example, polysiloxane- polycarbonate copolymers, polysiloxanepolyethylene oxide copolymers, polysiloxane
  • a biologically acceptable adhesive polymer matrix should be selected from polymers with glass transition temperatures below room temperature.
  • the polymer may, but need not necessarily, have a degree of crystallinity at room temperature.
  • Cross-linking monomeric units or sites can be inco ⁇ orated into such polymers.
  • cross-linking monomers can be inco ⁇ orated into polyacrylate polymers, which provide sites for cross-linking the matrix after dispersing the therapeutic agent into the polymer.
  • Known cross- linking monomers for polyacrylate polymers include polymethacrylic esters of polyols such as butylene diacrylate and dimethacrylate, trimethylol propane trimethacrylate and the like. Other monomers which provide such sites include allyl acrylate, allyl methacrylate, diallyl maleate and the like.
  • a plasticizer and/or humectant is dispersed within the adhesive polymer matrix.
  • Water-soluble polyols are generally suitable for this pu ⁇ ose. Inco ⁇ oration of a humectant in the formulation allows the dosage unit to absorb moisture on the surface of skin which in turn helps to reduce skin irritation and to prevent the adhesive polymer layer of the delivery system from failing.
  • Therapeutic agents released from a transdermal delivery system must be capable of penetrating each layer of skin. In order to increase the rate of permeation of a therapeutic agent, a transdermal drug delivery system must be able in particular to increase the permeability of the outermost layer of skin, the stratum corneum, which provides the most resistance to the penetration of molecules.
  • the fabrication of patches for transdermal delivery of therapeutic agents is well known to the art.
  • the therapeutic agents of the invention are conveniently delivered from an insufflator, nebulizer or a pressurized pack or other convenient means of delivering an aerosol spray.
  • Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the composition may take the form of a dry powder, for example, a powder mix of the therapeutic agent and a suitable powder base such as lactose or starch.
  • a powder mix of the therapeutic agent and a suitable powder base such as lactose or starch.
  • the powder composition may be presented in unit dosage form in, for example, capsules or cartridges, or, e.g., gelatine or blister packs from which the powder may be administered with the aid of an inhalator, insufflator or a metered-dose inhaler.
  • the therapeutic agent may be administered via nose drops, a liquid spray, such as via a plastic bottle atomizer or metered- dose inhaler.
  • atomizers are the Mistometer (Wintrop) and the Medihaler (Riker).
  • the local delivery of the therapeutic agents of the invention can also be by a variety of techniques which administer the agent at or near the site of disease.
  • site-specific or targeted local delivery techniques are not intended to be limiting but to be illustrative of the techniques available.
  • local delivery catheters such as an infusion or indwelling catheter, e.g., a needle infusion catheter, shunts and stents or other implantable devices, site specific carriers, direct injection, or direct applications.
  • the therapeutic agents may be formulated as is known in the art for direct application to a target area.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
  • the active ingredients can also be delivered via iontophoresis, e.g., as disclosed in U.S. Patent Nos.
  • the percent by weight of a therapeutic agent of the invention present in a topical formulation will depend on various factors, but generally will be from 0.01% to 95%> of the total weight of the formulation, and typically 0.1-25% by weight.
  • Drops such as eye drops or nose drops, may be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilizing agents or suspending agents.
  • Liquid sprays are conveniently delivered from pressurized packs. Drops can be delivered via a simple eye dropper-capped bottle, or via a plastic bottle adapted to deliver liquid contents dropwise, via a specially shaped closure.
  • the therapeutic agent may further be formulated for topical administration in the mouth or throat.
  • the active ingredients may be formulated as a lozenge further comprising a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the composition in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the composition of the present invention in a suitable liquid carrier.
  • a flavored base usually sucrose and acacia or tragacanth
  • pastilles comprising the composition in an inert base such as gelatin and glycerin or sucrose and acacia
  • mouthwashes comprising the composition of the present invention in a suitable liquid carrier.
  • formulations and compositions described herein may also contain other ingredients such as antimicrobial agents, or preservatives.
  • the active ingredients may also be used in combination with other therapeutic agents, for example, bronchodilators.
  • any physical or biological method that will introduce the vector into the muscle tissue of a host animal can be employed.
  • Vector means both a bare recombinant vector and vector DNA packaged into viral coat proteins, as is well known for AAV administration. Simply dissolving an AAV vector in phosphate buffered saline has been demonstrated to be sufficient to provide a vehicle useful for muscle tissue expression, and there are no known restrictions on the carriers or other components that can be coadministered with the vector (although compositions that degrade DNA should be avoided in the normal manner with vectors).
  • Pharmaceutical compositions can be prepared as injectable formulations or as topical formulations to be delivered to the muscles by transdermal transport. Numerous formulations for both intramuscular injection and transdermal transport have been previously developed and can be used in the practice of the invention.
  • the vectors can be used with any pharmaceutically acceptable carrier for ease of administration and handling.
  • solutions in an adjuvant such as sesame or peanut oil or in aqueous propylene glycol can be employed, as well as sterile aqueous solutions.
  • aqueous solutions can be buffered, if desired, and the liquid diluent first rendered isotonic with saline or glucose.
  • Solutions of the AAV vector as a free acid (DNA contains acidic phosphate groups) or a pharmacologically acceptable salt can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose.
  • a dispersion of AAV viral particles can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the sterile aqueous media employed are all readily obtainable by standard techniques well-known to those skilled in the art.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of a dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal and the like. In many cases it will be preferable to include isotonic agents, for example, sugars or sodium chloride.
  • Prolonged abso ⁇ tion of the injectable compositions can be brought about by use of agents delaying abso ⁇ tion, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by inco ⁇ orating the AAV vector in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by inco ⁇ orating the sterilized active ingredient into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and the freeze drying technique which yield a powder of the active ingredient plus any additional desired ingredient from the previously sterile-filtered solution thereof.
  • dilute sterile, aqueous solutions (usually in about 0.1 % to 5% concentration), otherwise similar to the above parenteral solutions, are prepared in containers suitable for inco ⁇ oration into a transdermal patch, and can include known carriers, such as pharmaceutical grade dimethylsulfoxide (DMSO).
  • DMSO dimethylsulfoxide
  • the therapeutic compounds of this invention may be administered to a mammal alone or in combination with pharmaceutically acceptable carriers.
  • the relative proportions of active ingredient and carrier are determined by the solubility and chemical nature of the compound, chosen route of administration and standard pharmaceutical practice.
  • the dosage of the present therapeutic agents which will be most suitable for prophylaxis or treatment will vary with the form of administration, the particular compound chosen and the physiological characteristics of the particular patient under treatment. Generally, small dosages will be used initially and, if necessary, will be increased by small increments until the optimum effect under the circumstances is reached. Exemplary dosages are set out in the example below.
  • the vectors of the invention may be employed to express a gene in any animal, and particularly in mammals, birds, fish, and reptiles, especially domesticated mammals and birds such as cattle, sheep, pigs, horses, dogs, cats, chickens, and turkeys. Both human and veterinary uses are particularly preferred.
  • the gene being expressed can be either a DNA segment encoding a protein, with whatever control elements (e.g., promoters, operators) are desired by the user, or a non-coding DNA segment, the transcription of which produces all or part of some RNA-containing molecule (such as a transcription control element, +RNA, or anti-sense molecule).
  • control elements e.g., promoters, operators
  • non-coding DNA segment the transcription of which produces all or part of some RNA-containing molecule (such as a transcription control element, +RNA, or anti-sense molecule).
  • Muscle tissue is a very attractive target for in vivo gene delivery and gene therapy, because it is not a vital organ and is very easy to access. If a disease is caused by a defective gene product which is required to be produced and/or secreted, such as hemophilia, diabetes and Gaucher' s disease, and the like, is muscle is a good candidate to supply the gene product if the appropriate gene can be effectively delivered into the cells.
  • Different vectors such as naked DNA, adenovirus and retrovirus, have been utilized to directly deliver various transgenes into muscle tissues. However, neither system can offer both high efficiency and long-term expression. For naked plasmid DNA directly delivered into muscle tissue, the efficiency is not high. There are only a few cells near the injection site that can maintain transgene expression.
  • the plasmid DNA in the cells remains as non-replicating episomes, i.e., in the unintegrated form. Therefore, it will be eventually lost.
  • adenovirus vector it can infect the non-dividing cells, and therefore, can be directly delivered into the mature tissues such as muscle.
  • the transgene delivered by adenovirus vectors are not useful to maintain long-term expression for the following reasons. First, since adenovirus vectors still retain most of the viral genes, they are not very safe. Moreover, the expression of those genes can cause the immune system to destroy the cells containing the vectors (see, for example, Yang et al. 1994, Proc. Natl. Acad. Sci. 91 :4407-4411).
  • adenovirus since adenovirus is not an integration virus, its DNA will eventually be diluted or degraded in the cells. Third, due to the immune response, adenovirus vector could not be repeatedly delivered. In the case of lifetime diseases, this will be a major limitation. For retrovirus vectors, although they can achieve stable integration into the host chromosomes, their use is very restricted because they can only infect dividing cells while a large majority of the muscle cells are non-dividing.
  • Adeno-associated virus vectors have certain advantages over the above- mentioned vector systems. First, like adenovirus, AAV can efficiently infect non-dividing cells. Second, all the AAV viral genes are eliminated in the vector. Since the viral-gene-expression-induced immune reaction is no longer a concern, AAV vectors are safer than Ad vectors. Thirds, AAV is an integration virus by nature, and integration into the host chromosome will stably maintain its transgene in the cells. Fourth, AAV is an extremely stable virus, which is resistant to many detergents, pH changes and heat (stable at 56°C for more than an hour). It can be lyophilized and redissolved without losing its activity. Therefore, it is a very promising delivery vehicle for gene therapy.
  • a recombinant AAV shuttle vector (AV.GFP3ori) which contained a GFP transgene cassette, bacterial ampicillin resistance gene, and bacterial origin of replication, was generated from a cw-acting plasmid (pCisAV.GFP3ori). Expression of the GFP gene was directed by the CMV promoter/enhancer and SV40 poly-adenylation sequences.
  • pCisAV.GFP3ori was constructed with pSub201 derived ITR elements (Samulski et al., 1987) and the intactness of ITR sequences was confirmed by restriction analysis with Smal and PvuII, and by sequencing.
  • Recombinant AAV stocks were generated by co-transfection of pCisAV.GFP3ori and pRep/Cap together with co-infection of recombinant Ad.CMVlacZ in 293 cells (Duan et al, 1997). Following transfection of forty 150 mm plates, cells were collected at 72 hours by centrifugation and resuspended in 12 ml of buffer (10 mM Tris pH 8.0). Virus was released from cells by three cycles of freeze/thawing and passaged through a 25 gauge needle six times.
  • Peak fractions of AAV were combined and re-purified through two more rounds of CsCl centrifugation, followed by heating at 58 °C for 60 minutes to inactivate all contaminant helper adenovirus.
  • this preparation gave approximate AAV titers of 10 12 DNA molecules/ml and 2.5 x 10 8 GFP-expressing units/ml.
  • Recombinant viral titers were assessed by slot blot and quantified against pCisAV.GFP3ori controls for DNA particles. Functional transducing units were quantified by GFP transgene expression in 293 cells.
  • helper adenovirus was confirmed by histochemical staining of rAAV infected 293 cells for beta-galactosidase, and no recombinant adenovirus was found in 10 10 particles of purified rAAV stocks.
  • the absence of significant wtAAV contamination was confirmed by immunocytochemical staining of rAAV/ Ad co-infected 293 cells with anti-Rep antibodies.
  • Zero hour controls were generated by mixing 10 9 particles of AV.GFP3ori with cell lysates prior to Hirt DNA preparation.
  • Hirt DNA isolated at each time point was used to transform E. coli SURE cells (Stratagene, La Jolla, CA.). Typically, 1/10 of the Hirt DNA preparation was used to transform 40 ml of competent bacteria by electroporation. The resultant total number of bacterial colonies was quantified for each time point and the structure of circular intermediates was evaluated for greater than 20 plasmid clones for each time point from two independent experiments. Structural determinations were based on restriction enzyme analysis using Pstl, Sphl, Asel single and double digests together with Southern blotting against GFP, stuffer, and ITR probes. Evaluation of E2a and GFP gene expression in Hela cells.
  • Hela cells were harvested at 24 or 72 hours post-infection by trypsinization, resuspended in 2%>FCS/PBS and evaluated by FACS analyses. Thresholds were set using uninfected controls and the percentage and/or the average relative fluorescent intensity was determined by sorting greater than 10 5 cells per experiment condition.
  • AV.GFP3ori virus ( Figure IB) was generated from a cw-acting plasmid (pCisAV.GFP3ori, Figure IA) by co-transfection in 293 cells with trans-acting plasmids encoding Rep and Cap viral genes.
  • This viral vector (AV.GFP3ori) encoded the green fluorescent protein (GFP) reporter gene, a bacterial origin of replication (ori), and the bacterial ampicillin-resistance gene.
  • Ori and ampicillin-resistance sequences encoded in this virus allow for the rescue of circular AAV genomes formed during the transduction process.
  • helper adenovirus Many aspects of the wtAAV growth cycle are affected by helper adenovirus, including AAV DNA replication, transcription, splicing, translation, and virion assembly.
  • helper adenovirus including AAV DNA replication, transcription, splicing, translation, and virion assembly.
  • Ad early gene products provide helper functions for the wtAAV lytic cycle, including: Ela, Elb, E2a, E4 ORF6 and VAI RNA (Muzyczka, 1992).
  • one of the most critical factors which is required for AAV replication is the 34 kD E4 protein (ORF6).
  • Ad E4 ORF6 is essential for the augmentation of rAAV transgene expression seen with adenovirus co-infection (Ferrari et al., 1996; Fisher et al., 1996). According to these reports, the rate-limiting step enhanced by these adenoviral proteins is the conversion of single stranded AAV genomes to double stranded forms.
  • retroviral transduction intermediates have striking similarities to the current findings with AAV.
  • Three DNA forms have been isolated following retroviral infection, including linear DNA with long terminal repeats (LTRs) at both ends, circular DNA with one LTR, and circular DNA with multiple LTRs (Panganiban, 1985).
  • LTRs long terminal repeats
  • Cirlusaniban a DNA with multiple LTRs
  • AAV circular intermediates The head-to-tail ITR structures found in AAV circular intermediates are most characteristic of latent integrated AAV genomes. In contrast, lytic phases of AAV growth are typically associated with head-to-head and tail-to-tail replication form genomes. Hence, it is likely that circular intermediates represent a latent aspect of the AAV life cycle. The finding that co-infection with adenovirus leads to increased abundance and stability of AAV circular intermediates suggests a novel link between adenoviral helper functions and latent infection of AAV.
  • inverted head-to-tail ITRs which include palindromic hai ⁇ ins similar in structure to "Holliday-like" junctions, might impart recombinagenic activity which aids in viral integration.
  • Holliday junctions have been shown to play critical roles in directing homologous recombination in bacteria through the processing of recombination intermediates by RuvABC proteins (West, 1997; Lee et al., 1998).
  • RuvABC proteins West, 1997; Lee et al., 1998.
  • a mammalian endonuclease analogous to bacterial RuvC resolvase, has also been isolated from cell lines (Hyde et al., 1994).
  • the cts-acting plasmid (pCisAV.GFP3ori) used for rAAV production was generated by subcloning the Bsp 1201 /Not I fragment (743 bp) of the GFP transgene from pEGFP-1 (Clontech) between the CMV enhancer/promoter and SV40polyA by blunt-end ligation.
  • a 2.5 kb cassette containing beta-lactamase and bacterial replication origin from pUC19 was blunt ligated down-stream of GFP reporter cassette.
  • the ITR elements were derived from pSub201.2
  • the entire plasmid contains a 4.7 kb AAV component flanked by a 2 kb stuffer sequence.
  • ITR sequences were confirmed by restriction analysis with Smal and PvuII, and by direct sequencing using a modified di-deoxy procedure which allowed for complete sequence through both 5' and 3' ITRs.
  • Recombinant AAV stocks were generated by co-transfection of pCisAV.GFP3ori and pRep/Cap together with co-infection of recombinant Ad.CMVlacZ in 293 cells.
  • the rAV.GFP3ori virus was subsequently purified through 3 rounds of CsCl banding as described in Duan et al., 1997. The typical yields from this viral preparation were 1012 DNA molecules/ml.
  • DNA titers were determined by viral DNA slot blot hybridization against GFP 32 P-labeled probe with copy number plasmid standards.
  • the absence of helper adenovirus was confirmed by histochemical staining of rAAV infected 293 cells for beta-galactosidase, and no recombinant adenovirus was found in 10 10 particles of purified rAAV stocks.
  • the absence of significant wtAAV contamination was confirmed by immunocytochemical staining of rAAV/ Ad co-infected 293 cells with anti-Rep antibodies. Transfection with pRep/Cap was used to confirm the specificity of immunocytochemical staining. No immunoreactive Rep staining was observed in 293 cells infected with 10 10 rAAV particles.
  • the tibialis anterior muscle of 4-5 week old C57BL/6 mice were infected with AV.GFP3ori (3 X 10 10 particles) in Hepes buffered saline (30 ⁇ l).
  • GFP expression was analyzed by direct immunofluorescence of freshly excised tissues and/or in formalin-fixed cryopreserved tissue sections in four independently injected muscles harvested at 0, 5, 10, 16, 22 and 80 days post-infection. Tissue sections were counter-stained with propidium iodide to identify nuclear DNA.
  • Hirt DNA Hirt, 1967 (20 ml per muscle sample) was isolated from at least three independent muscle specimen for each time point and used to transform E.
  • a Purified virus was reconstituted into muscle homogenates prior to preparation of Hirt DNA.
  • Viral DNA predominantly contained single stranded genomes as evident by Southern blot analysis against with ITR probe. However, small amount of dsDNA AAV genomes also existed and are likely due to reannealing of single stranded genomes during preparation. Purified viral DNA concentrations were determined by OD 260 and 75 ng representing approximately 3 x 10 10 viral genomes were used for transformation of bacteria.
  • Hindlll/PvuII digestion was used to remove the entire rAAV genome from pcisAV.GFP3ori. Hindlll and PvuII leave 10 and 0 bps of flanking sequence outside the 5' and 3' ITRs, respectively.
  • the linear dsDNA fragment (4.7 kb) was gel isolated following blunting with T4 DNA polymerase and the DNA concentration determined by OD 260 . One hundred and fifty ng of linear fragment representing approximately 3 x 10 10 viral genomes were used for transformation of bacteria.
  • d Linear dsDNA viral genomes (Hindlll/PvuII blunted fragment) were treated with T4 DNA ligase prior to transformation of bacteria.
  • Preparative-scale fractionation of the muscle Hirt DNA was performed by 1% agarose gel electrophoresis using the Bio-Rad Mini Prep Cell (Catalog #170-2908).
  • a 4.5 ml (10.5 cm) tubular gel containing 1 x TBE buffer was poured according to manufacturer's specification.
  • a total of 20 ml Hirt preparation from one entire muscle sample was loaded on top of the gel.
  • Electrophoresis was carried out at a constant current of 10 mA over a period of 5 hours.
  • Sample eluent was drawn from the preparative gel apparatus by a peristaltic pump at a rate of 100 ml/min and eluted into a fraction collector at 250 ml/fraction.
  • the collected DNA was subsequently concentrated by standard ethanol precipitation and used to transform SURE bacterial cells by electroporation as described above. In vitro Persistence of AAV Circular Intermediates.
  • AAV circular intermediate plasmid clones were evaluated following transient transfection in Hela and 293 cells.
  • Subconfluent monolayers of Hela cells in 24-well dishes were transfected with 0.5 mg of either AAV circular intermediates (p81 or p87) or pCMVGFP using Lipofectamine (Gibco BRL Inc.). The cultures were then incubated for 5 hours in serum free DMEM followed by incubation in DMEM supplemented with 10%) fetal bovine serum. All plasmid DNA samples used for transfections were spiked with pRSVlacZ (0.5 mg) as an internal control for transfection efficiency.
  • the head-to-tail ITR DNA element was subcloned into the pGL3 luciferase plasmid to generate pGL3(ITR).
  • the head-to-tail ITR DNA element was isolated from a monomer circular intermediate (p81) by Aatll and Haell double digestion and subsequently inserted into the Sail site of pGL3 (Promega) by blunt ligation.
  • the resultant plasmid pGL3(ITR) contains the luciferase reporter and head-to-tail ITR element 3 ' to the polyA site. The integrity of the ITR DNA element within this plasmid was confirmed by sequencing.
  • AAV Circular Intermediates Represent Stable Episomal Forms of Viral DNA Associated with Long-term Persistence of Transgene Expression in Muscle.
  • AV.GFP3ori a rAAV shuttle viral vector which harbors an ampicillin resistance gene, bacterial origin of replication, and GFP reporter gene ( Figure 1 A). This recombinant virus was used to evaluate the presence of circular intermediates by bacterial rescue of replication competent plasmids.
  • Figure 5B demonstrates several isolated circular intermediates with 1-3 ITRs isolated from 80 days muscle Hirt samples. This is in contrast to the more uniform structure of circular intermediates with two ITRs in a head-to-tail conformation at 5-22 days post-infection.
  • control experiments were performed. First, uninfected control muscle Hirt preparations, spiked with an equal amount of rAAV virus used for in vivo infection of muscles, failed to give rise to replicating plasmids following transformation of E.coli.
  • Hirt used for transformation was 3/20 the entire Hirt DNA.
  • the numbers have been adjusted to reflect viral innoculum and yields for the entire muscle.
  • Plasmid DNA was spiked into mock infected muscle homogenates prior to isolation of Hirt DNA. This reconstituted Hirt DNA was then used for transformation of bacteria.
  • Control LacZ plasmid was approximately 7000 bp with a molecular weight of 4.6 x 10 6 g/mole.
  • AAV Circular Intermediates Demonstrate Increased Persistence as Plasmid Based Vectors.
  • rAAV circular head-to-tail intermediates may be molecular structures of the AAV genome associated with the latent life cycle and increased episomal stability.
  • Several aspects of the structure of AAV circular intermediates may account for their increased stability in vivo.
  • circularization of AAV genomes may create a nuclease resistant conformation.
  • these sequences might bind cellular factors capable of stabilizing these structures in vivo.
  • AAV circular intermediates have increased persistence in cell lines in vitro, lends support to the hypothesis that these structures represent stable episomal forms following rAAV transduction in muscle. Stability of circular intermediates in vivo might be mediated by the binding of cellular factors to "Holliday-like" junctions in ITR arrays which stabilize or protect DNA from degradation.
  • rAAV has been shown to be an efficient vector for expressing transgenes in various tissues in addition to muscle, such as brain, retina, liver, lung, and hematopoetic cells (Snyder et al., 1997; Muzyczka, 1992; Kaplitt et al., 1994; Walsh et al, 1994; Halbert et al., 1997; Koeberl et al., 1997; Conrad et al., 1996; Bennett et al., 1997; Flannery et al., 1997).
  • rAAV the mechanisms of in vivo rAAV-mediated transduction and persistence of transgene expression still remain unclear.
  • Persistence of AAV circular intermediates were assessed by injection of plasmid DNA directly into the pronucleus of fertilized Xenopus oocytes. Twenty- five ng of the p81 isolate of AAV circular intermediates was injected at the single cell stage of fertilized Xenopus oocytes. This plasmid was compared to the proviral plasmid pCisAV.GFP3ori, which contains two ITRs separated by stuffer sequence in an alternative confirmation to ITRs in p81.
  • Figure 13 depicts the persistence of GFP plasmids as assessed by direct fluorescence of GFP. At this state of tadpole development, the fertilized oocyte has expanded from a single cell to approximately 10 6 cells.
  • AAV circular intermediates confer a higher level of stability in development Xenopus oocytes than plasmids containing similar transcriptional elements and ITR sequences in an alternative confirmation. Given that in the case of p81 injected oocytes, tadpoles are completely fluorescent, the data suggests that some level of integration may have occurred.
  • AAV adeno-associated virus
  • AAV circular intermediates correlates with the onset and maintenance (at 80 days) of transgene expression, respectively.
  • a 300 bp fragment encompassing the head-to-tail inverted ITR repeats found in AAV circular intermediates when cloned into heterologous expression plasmids can confer increased stability to those plasmids in HeLa cells.
  • the structural aspects of AAV circular intermediates may lead to development of non- viral, plasmid based, gene transfer vectors with increased persistence of transgene expression.
  • AAV circular intermediates which differ in length and/or sequence of the ITR array are more efficacious plasmid based vectors for liposome-mediated gene transfer to the airway and muscle
  • several distinct forms of AAV circular intermediates are evaluated as plasmid-based delivery systems in three model systems of the airway including: 1) in vitro polarized primary airway epithelial monolayers, 2) mouse lung, and 3) human bronchial xenografts.
  • Persistence is evaluated at both the level of transgene expression (using GFP and luciferase reporters) and at the level of episomal and integrated transgene derived DNA. Studies are performed to assess whether integration can be specifically enhanced by co-transfection with Rep DNA or mRNA. These studies also evaluate both the extent of integration and site specificity to AVS1 sites in chromosome 19 of human model systems.
  • AAV circular intermediates which confer increased persistence of transgene expression include a DNA element encompassing the head-to-tail ITR. Based on the findings that circular intermediates have increased episomal persistence in muscle following rAAV transduction, these structures may also have increased persistence as plasmid-based vehicles to the airway. Interestingly, several naturally occurring mutations which are found in approximately 50%> of AAV circular intermediates affect the stability of the intermediate.
  • exogenously supplied E2a DNA binding protein may also enhance stability of AAV circular intermediates.
  • Rep may increase the integration of circular intermediates while E2a may increase their episomal stability.
  • E2a DBP DNA binding protein
  • its encoded nuclear localization sequence NLS
  • E2a may act to alter the persistence of AAV circular intermediates through the induction of cellular factors which interact with the ITR array.
  • Liposome mediated gene transfer to the airway has considerable advantages due to the low level of toxicity.
  • limitations include transient low level expression in differentiated airway epithelia.
  • several laboratories have had considerable success with the use of cationic liposome-mediated gene transfer in several animal models including mouse and rat lung, and numerous laboratories have pursued clinical trials, which suggested that these vehicles may show promise for gene therapy of the cystic fibrosis (CF) lung.
  • delivery of the present vectors in plasmid form via liposomes may be a safe and effective vehicle for gene transfer to the airway.
  • AAV circular intermediates may also have increased persistence in airway epithelial cells as seen in Hela cells.
  • several distinct forms of circular intermediates delivered by liposome-mediated transfection into primary airway epithelial cells are evaluated. Based on the diversity of ITR repeat elements between various isolated circular intermediates (i.e., including 0, 1, 2, and 3 ITRs), circular intermediates isolated from later time points in muscle may have been naturally selected for increased stability in vivo.
  • ITR repeat elements between various isolated circular intermediates i.e., including 0, 1, 2, and 3 ITRs
  • ITRs are transfected into primary airway cultures and polarized epithelial cell monolayers using the cationic lipid GL-67 (Genzyme Inc.). DNA to lipid ratios are optimized using a luciferase reporter. Additionally, the addition of EGTA, or the use of calcium-free media, can increase the extent of gene transfer about 10- fold, and may be included to enhance gene transfer to polarized epithelial ' monolayers. To evaluate persistence and expression of transgenes from circular intermediates, direct fluorescent microscopy and Southern blotting of both Hirt and genomic DNA with GFP P 32 -labeled probes are utilized. Proliferating cultures of primary airway epithelial cells can be passaged up to 4 times during this analysis.
  • polarized epithelial monolayers are evaluated at 1 week intervals for DNA persistence for up to 6 weeks. Since GFP transgene expression may be low and difficult to detect by direct fluorescence, GFP is quantitated by fluorometer of cell lysates.
  • circular intermediates may form within cells and certain structures of these intermediates may persist by virtue of affinity for cellular factors which bind at ITR arrays. If this is true, then it may be possible to select for and isolate optimal circular intermediates with increased persistence in airway cells by batch screening of circular intermediates pools from rAAV infected airway epithelia. Primary airway epithelia cell cultures are infected with AV. GFP3ori
  • Hirt DNA containing circular intermediates from rAAV infected cells is used to then transfect primary airway epithelial cells from which Hirt DNA is prepared at 5-15 days post-transfection.
  • This second Hirt isolation is then used to isolate replication competent plasmids following transformation into bacteria. This selection process may give rise to those populations of circular intermediates with increased episomal persistence in airway epithelial cells. Selected clones of circular intermediate plasmids isolated by this procedure are then tested individually for increased persistence following liposome mediated transfection. These studies are performed in a batch type screening in 24 well plates using two serial passages for persistence.
  • plasmids having increased persistence are isolated, their structure and sequence of ITR arrays are characterized. Since screening is performed on small-scale cultures, it may be necessary to implement semi-quantitative screening for DNA persistence within the first round of transfection using PCR methods. Candidate plasmids with a high level of increased persistence as compared to control plasmids which lack ITR sequences but contain the identical promoter-reporter element, are evaluated on a larger scale transfection amenable to analysis by Southern blotting of total DNA.
  • mice are transfected with GL-67/DNA complexes at a ratio of 25 ⁇ g plasmid/25 ⁇ g lipid in an iso-osmotic solution of Dextrose.
  • mice are harvested for immunofluorescent detection of GFP in formalin fixed sections and for quantitative fluorometry of tissue lysates.
  • Southern blots are employed to evaluate the persistence of plasmids in Hirt and genomic DNA.
  • luciferase constructs are evaluated in which the ITR array has been cloned either 5 ' or 3 ' to the reporter gene. Furthermore, the use of luciferase reporters allows for more sensitive assessment of transgene activity in cell lysates. Similarly, in vivo persistence of transfected circular intermediates and heterologous plasmids containing ITR arrays found within circular intermediates is evaluated in human bronchial xenografts.
  • E2a DBP leads to a 10-fold increase in the abundance of circular intermediates as compared to E2 deleted virus.
  • studies with El -deleted virus have demonstrated that the persistence of circular intermediates in Hela cells is increased at 72 hours post-infection.
  • E2a DBP may augment circular intermediate formation and/or increase the stability of these structures by an unknown mechanism.
  • E2a DBP may interact directly with circularized genomes and/or induce cellular factors which interact with sequences in these AAV genomes. Since DBP encodes an NLS, this protein may act to shuttle circular intermediates to regions of nucleus that allow for increased stability of these structures.
  • NLS sequences have been shown to cooperatively interact with nucleolar targeting sequences and hence we will also evaluate if subnuclear targeting is important in maintaining the increased stability of circular intermediates containing ITR arrays. Furthermore, it is currently unknown where circular intermediates form in the cell and it remains plausible that they may form in the cytoplasm or nucleus. Hence, if DBP associates directly with circular intermediates, it may act as an NLS for DNA to enter the nucleus as well.
  • Several in vitro reconstitution models are used to investigate the interaction of circular intermediates with DBP and their affect on in vivo persistence following DNA transfection in Hela cells.
  • gmDBP6 Hela cell line
  • This cell line gives rise to high levels of DBP in nuclear extracts by Western blot following treatment with dexamethasone.
  • gmDBP ⁇ cells (+/- DEX) are transfected with various AAV circular intermediate plasmids containing 0, 1, 2, and 3 ITRs and total cellular and nuclear plasmid content evaluated by subcellular fractionation using Southern blotting against GFP probes. The time course of these studies is initially within the range of 12 hours to 4 days post-transfection.
  • Transgene expression is evaluated by fluorometry (in cell lysates), and fluorescent microscopy (in viable cells), for GFP and luminescence for luciferase.
  • fluorometry in cell lysates
  • fluorescent microscopy in viable cells
  • GFP and luminescence for luciferase.
  • Hela cells have demonstrated that immediate increases in transgene expression from AAV GFP circular intermediates as compared to control GFP plasmids occur as early as 24 hours post-transfection.
  • certain cellular factors may facilitate an immediate accumulation of circular intermediates in the nucleus. DBP may invoke this increase by either direct interactions with ITR sequences or by the induction of cellular factors.
  • microinjection experiments in oocytes are performed with 50 ng of plasmid DNA of circular intermediates with and without 50 ng of DBP mRNA.
  • Experiments initially evaluate the time course of GFP transgene expression (+/- DBP cRNA) by direct fluorescent microscopy. If major differences are seen, quantitative fluorometry of individual whole oocytes in 96 well plates is conducted. Similar studies on nuclear targeting in the presence of DBP can also be evaluated in this model by pooling microinjected oocytes for nuclear isolation and Southern blot analysis.
  • a third experimental model to evaluate nuclear targeting and/or accumulation of circular intermediate vectors in the presence and absence of DBP involves the microinjection of fluorescently labeled plasmid DNA into the cytoplasm and real time imaging to follow the nuclear accumulation of DNA.
  • the DNA fluorescent dye, TOTO-1 is used to label DNA prior to injection. This dye forms an extremely stable complex with negligible diffusion and re- inco ⁇ oration into nuclear DNA following transfection into polarized airway epithelial cell monolayers.
  • Co-localization of DBP with wtAAV DNA genomes at focal hot spots within the nucleus supports the observation that nucleolar targeting may be important for persistence.
  • the extent of integration is also evaluated by two criteria, Southern blotting of Hirt and genomic DNA and clonal expansion of GFP expressing cells. Since Southern blot has an approximate limit of sensitivity of 1 integrated plasmid molecule per 10 cellular genomes, clonal expansion may be necessary to evaluate persistence in less transfectable cells such as CFT1 and IB-3 cells. Cell lines are evaluated over the course of 1-10 passages.
  • synthetic DNA sequences are generated with identical secondary structure to several ITR arrays in circular intermediates.
  • the primary sequence is completely altered and bares no resemblance to sequences contained within native AAV ITRs.
  • These synthetic DNA sequences are tested for their ability to confer increased episomal stability to heterologous plasmids in several model systems including: 1) the airway, 2) muscle, 3) and developing Xenopus embryos.
  • the developing Xenopus embryo model is ideal for testing integration and persistence of plasmid based vectors for application of in utero gene therapy.
  • AV.GFP3ori (Example 1) and AVAlkphos (also known as CWRAPSP, a gift of Dusty Miller) (Halbert et al., 1997).
  • Virus stocks were generated by co- transfection of 293 cells with either pCisAV.GFP3ori or pCWRAPSP along with pRep/Cap, followed by co-infection with recombinant Ad.CMVlacZ helper virus (Example 2).
  • rAAV was then purified through three rounds of CsCl density gradient centrifugation as previously described by Duan et al. (1997). Purified viral fractions were heated at 60°C for 1 hour to inactivate any residual contaminating helper adenovirus.
  • AV.GFP3ori and AVAlkphos were 1 x 10 12 and 7 x 10 u particles per ml, respectively, as determined by slot blot hybridization with 32 P-labeled GFP or Alkphos probes.
  • Infectious titers determined by infection of 293 cells with rAAVs were 1.1 x 10 9 IU/ml (AV.GFP3ori) or 8.6 x 10 8 IU/ml (AVAlkphos).
  • Controls testing for contamination of rAAV stocks with wtAAV by anti-Rep immunocytochemical staining in rAAV/ Ad.CMVlacZ co-infected 293 cells were negative (limit of sensitivity is less than 1 infectious wtAAV particle per 10 10 DNA particles of rAAV).
  • histochemical staining for ⁇ -galactosidase in rAAV infected 293 cells showed no detectable contamination with helper adenovirus in 10 10 DNA particles of rAAV (limit of sensitivity). Infection of muscle tissue and evaluation of transgene expression.
  • mice used for these experiments were housed in a virus- free animal care facility and were maintained under strict University of Iowa and NTH guidelines, using a protocol approved by the Animal Care and Use Committee and facility veterinarians.
  • Four to five week old mice received bilateral 30 ⁇ l injections of a mixture of both AV.GFP3ori and AVAlkphos into the tibialis anterior muscle (5 x 10 9 DNA particles of each virus per muscle).
  • Controls included uninjected muscles and muscles receiving injections ofone of the viruses alone. At 14, 35, 80, and 120 days post-infection, animals were euthanized and tissues were harvested for evaluation of transgene expression and preparation of low molecular weight Hirt DNA. For each experimental time point, at least 3 independently injected muscles were evaluated.
  • the percentages of Alkphos and/or GFP hybridizing plasmids were calculated by this method for each muscle sample. From this percentage, the total number of plasmids hybridizing to each probe in the Hirt DNA sample was calculated from the total CFU obtained in each transformation. In this analysis, each muscle sample was evaluated independently to determine the mean (+/-SEM) total Alkphos and/or GFP hybridizing plasmids. A second evaluation involved the transfection of rescued plasmids into 293 cells using lipofectamine, followed by evaluation of GFP fluorescence and histochemical staining for Alkphos.
  • plasmid structure was mapped by Southern blotting and restriction enzyme analysis. The structural of five co- expressing circular intermediate plasmids were determined by digestion with Ahdl, Hindlll, Notl, Hindlll/Notl, Clal/Asel, and/or SnaBI and Southern blotting was performed with 32 P-labeled GFP, Alkphos, and ITR probes.
  • the tibialis anterior muscle of mice was co-infected with 5 x 10 9 DNA particles of both AV.GFP3ori and AV.Alkphos.
  • muscles were harvested and analyzed for transgene expression.
  • Transgene expression from both reporters was weak but clearly visible in 14 day muscle samples.
  • transgene expression was maximal and serial sections demonstrated expression of both Alkphos and GFP transgenes in overlapping regions of the muscle ( Figures 15A- C). At this time point, approximately 50% of the fibers in the tibialis muscle expressed both transgenes.
  • GFP/ Alkphos co-hybridizing plasmids were never observed.
  • the percentage of GFP/Alkphos co-hybridizing plasmids increased with time and reached 33% by 120 days (Figure 16C).
  • Intermolecular recombination of rAAV genomes to form single circular episomes may be particularly useful for gene therapy.
  • large regulatory elements and genes beyond the packaging capacity of rAAV may become linked after co-infecting tissue with two independent vectors ( Figure 19).
  • This strategy could also involve trans-splicing vectors encoding two independent regions of a gene which are brought together to form an intact splicing unit by circular concatamerization.
  • the pcisAV.Luc proviral plasmid was generated by cloning the 1983 bp Nhel/BamHI fragment from pGL3-Basic (Promega), containing the luciferase gene and SV40 late polyA signal, by blunt-end ligation into the blunted Xba I site of pSub201 (Samulski et al., 1987).
  • pcisAV.SV(P)Luc was generated using a blunted 2175 bp Nhel/BamHI fragment, from the pGL3- Promoter (Promega), containing the SV40 promoter, luciferase gene, and SV40 late polyA signal.
  • the pcisAV.SV(P/E)Luc plasmid was generated by blunt- end hgation of a 2427 bp Nhel/Sall fragment from pGL3-Control (Promega) into the blunted Xba I site of psub201.
  • This construct contains the SV40 promoter, luciferase gene, SV40 late polyA signal and SV40 enhancer.
  • the "super-enhancer" vector, pcisAV.SupEnh, was produced using a two-step cloning process. First, a 0.62 kb blunted Bglll/Pvul fragment containing the CMV immediate early enhancer from pIRES (Clontech) was subcloned into the blunted BamHI site in pGL3-Control (Promega) to make pGL3-Control-CMVenh.
  • pcisAV.SupEnh contains the SV40 enhancer, the CMV immediate early enhancer, the ⁇ -lactamase gene, and a bacterial replication origin.
  • the ampicillin resistance gene ( ⁇ -lactamase) and bacterial original of replication were included in pcisAV.SupEnh to facilitate the subsequent rescue of circular AAV genomes from infected cells in bacteria.
  • the control vector, pcisAV.AmpOri was generated by blunt-end ligation of a 1.1 kb Sail digested stuffer sequence from the human glycosylasparaginase cDNA into Pstl digested pcisAV.GFP3ori.
  • This plasmid has a structure similar to that of pcisAV.SupEnh, except that it does not contain any enhancer elements.
  • the pcisAV.AmpOri was used as a negative control for non-specific enhancement of transgene expression by intermolecular recombination of two different AAV vectors.
  • ITR sequences in all the plasmids were confirmed by digestion with restriction enzymes, including Smal, Mscl, and BssHII, which have unique cutting sites within different regions of ITR. All the viral preparations were obtained according to a method described in Duan et al. (1997). The quality of the viral stocks (i.e., contamination with adenovirus and/or wild type AAV) was confirmed as previously described in Duan et al. (1998b). The analyses showed less than 1 recombinant adenovirus and wt AAV infectious particles per 10 10 particles of rAAV. Viral titers were determined by quantitative slot-blot hybridization using either luciferase, CMV enhancer, SV40 enhancer, or ori probes for each of the respective vectors against plasmid copy number standards.
  • Luciferase assays were performed from cell lysates harvested from either in vttro-infected human fibroblasts or from in vo-infected mouse tibialis anterior muscle. Human fibroblasts were infected with virus in 60 mm dishes. These in tro-infected cells were harvested at 3 days post-infection by rinsing cells with PBS twice, and then incubating with lx Report lysis buffer (Promega) (400 ⁇ l per 60 mm plate) at room temperature for 15 minutes. Cells were scraped into an eppendorf tube and centrifuged for 30 seconds at 14,000 ⁇ m.
  • the entire muscle was harvested at 30 days or 90 days post-infection and weighed prior to cell lysate preparation.
  • the muscle tissue was frozen in liquid nitrogen and pulverized by hand grinding with an ice-cold porcelain mortar and pestle.
  • the muscle was further minced and homogenized in 100 ⁇ l of lx Report lysis buffer with a hand-held plastic pestle for 2 minutes (Kontes, Vineland, New Jersey). After 15 minutes incubation at room temperature, the crude lysates were spun for 30 seconds at 14,000 ⁇ m, and the supernatants were used for luciferase activity assay as described above. To minimize variability, all experimental samples were analyzed simultaneously using the same batch of luciferase assay reagents and were normalized to the protein content in the lysate. Results
  • Co-administration of a cts-activating vector increases rAAV mediated luciferase expression in fibroblasts.
  • AV.Luc contains the luciferase transgene and an SV40 poly A signal but no promoter sequences.
  • AV.SV(P)Luc is similar to AV.Luc except that an SV40 promoter (lacking the enhancer sequence) was inserted in front of the luciferase transgene.
  • AV.SV(P/E)Luc includes both the SV40 promoter and enhancer, driving expression of the luciferase transgene, and was used as a control for maximal expression in the absence of intermolecular recombination with an enhancer containing vector.
  • an rAAV "super- enhancer" vector (AV.SupEnh) was also constructed, which contains SV40 and CMV enhancer regions without promoter or transgene sequences.
  • a negative control vector (AV.AmpOri) which is similar to AV.SupEnh except that the enhancer sequences were replaced by a non-specific stuffer fragment, was also constructed.
  • Enhancers are cz ' s-acting DNA sequences that can be recognized by regulatory proteins to stimulate transcription in a context independent manner relative to the promoter and transgene. If cells were co-infected with AV.SV(P)Luc and AV.SupEnh vectors, luciferase transgene expression could be significantly increased from the minimal SV40 promoter only if intermolecular recombination had occurred between the two independent vectors ( Figure 21). However, in accordance with the definition of an enhancer, no activation should occur if the enhancer sequences and the transgene cassette are located in separate circular DNA molecules (Lewin, 1997).
  • Hirt DNA from infected cells was transformed into competent SURE E. Coli cells.
  • no bacterial clones were retrieved from cells infected with either AV.Luc or AV.SV(P)Luc alone (neither vector contains amp r and ori sequences).
  • AV.SupEnh which contains amp r and ori sequences but no luciferase gene
  • approximately 4%> of the rescued clones also contained the luciferase transgene, according to restriction enzyme mapping and Southern blotting analyses.
  • the bacterially rescued concatamers containing both the luciferase transgene and AV.SupEnh also demonstrated greater than a 100-fold higher luciferase activity than the original proviral luciferase plasmid (pcisAV.Luc) alone.
  • 90 day muscles infected with 2 x 10 10 particles of the control AV.SV(P/E)Luc vector (which contains both the SV40 enhancer and promoter) produced luciferase levels that were 10- and 100-fold less than the levels seen following co-infection with AV.SupEnh/ AV.Luc and AV.SupEnh AV.SV(P)Luc, respectively.
  • rAAV transduction, circularization and concatamerization can be used to deliver CFTR transgene cassettes to the airway.
  • Two approaches may be employed in which various genetic elements of the CFTR transgene cassette are split into two or more vectors which are then used for co-infection of the airway.
  • trans-splicing utilizes a rAAV vector harboring the promoter/enhancer driving the first half of CFTR DNA flanked by a donor splice site (vector- 1, Figure 19C) and a second rAAV vector harboring the second half of CFTR DNA and a polyA addition sequence preceded by a donor acceptor site (vector-2, Figure 19C).
  • a second approach, “cts-activation” employs a first vector that harbors the entire CFTR transgene with a minimal synthetic promoter and a second vector comprises several strong enhancer sequences. Through the process of concatamerization, these two vectors are brought into juxtaposition with one another, allowing for splicing or cts-activation of enhancer/promoter combinations.
  • Assays for splicing integrity include RNase protection assays of transfected MDCK cells for sequences within the large T antigen intron and surrounding regions. Polarized airway epithelial cells grown at the air-liquid interface are co-infected with the donor and acceptor CFTR AAV vectors and CFTR gene expression in these cells is then monitored by both immunofluorescent localization and functional analysis of short circuit currents (Smith et al., 1992; Smith et al., 1990). Additionally, functional assays using two electrode voltage clamp measurements (TEV) of oocytes following nuclear microinjection of the chimeric plasmid are used to demonstrate intact splicing and active CFTR channels (Zhang et al,
  • Hirt analyses of episomal AAV species are used to correlate the efficacy and persistence of CFTR gene expression with the formation of AAV circular intermediates.
  • CFTR expressing circular concatamers Several systems are utilized to evaluate the efficiency of intermolecular recombination to form functional CFTR expressing circular concatamers.
  • the first employs short circuit measurements in polarized CF airway epithelia.
  • Conditions for rAAV infection include basolateral infection of polarized CF epithelia, preferably in the presence of agents that enhance the level of transgene expression.
  • a second model employs CF human bronchial xenografts to evaluate CFTR function complementation.
  • Reconstitution of xenografts with UV-irradiated rAAV infected primary cells can resulted in approximately 50% transduction in differentiated xenografts.
  • - Functional expression of CFTR in CF primary airways is evaluated by transepithelial potential difference (PD) measurements (Jiang et al., 1998).
  • a "czs-activation" approach may be employed (Example 6) to utilize the concatamerization process to deliver full-length CFTR.
  • a synthetic minimal promoter driving full-length CFTR within one viral vector and a second independent vector encoding a tandem array of strong enhancer sequences such as RSV, CMV, and SV40 may be employed.
  • a trans-splicing approach was employed to deliver the genomic epo gene to mice.
  • the vectors are shown in Figure 25.
  • the infection of mice with these vectors protected the mice from adenine induced anemia brought on by renal failure.
  • a rAAV vector comprising a replication origin of a circular episome is employed.
  • a rAAV vector comprising the EBV replication origin (OriP) and EBNA-1, the only viral protein needed to facilitate replication at this origin is prepared (Vector 2, Figure 24).
  • DNA fragments encoding EBNA-1 and OriP are excised from pREPIO (Clontech Inc.) and cloned into a pCisAV.RSV vector.
  • virus is produced from pCisAV.EBNA-1/OriP construct and a reporter construct, e.g., AV.GFP3ori, and virus is used to co-infect Hela cells.
  • Transgene expression is quantitated following each passage and Hirt DNA is isolated for Southern blot analysis of episomal DNA persistence. If persistence of GFP expression is increased in the presence of co-infection with EBNA-1 rAAV vectors following serial passage, there will be an increased abundance of co-encoding EBNA-1 /GFP rescued plasmids with increasing passage number.
  • human bronchial xenografts and the RSV-EBNA-1 rAAV vector and a second CFTR rAAV vector with a minimal promoter may be employed.
  • the RSV enhancer may increase transcription from the minimal CFTR promoter.
  • J.M. Transduction with recombinant adeno-associated virus for gene threapy is limited by leading-strand synthesis. J. Virol. 70, 520-532 (1996).
  • Fisher K. J. et al. Recombinant adeno-associated virus for muscle directed gene therapy. Nat. Med. 3, 306-312 (1997). Fisher-Adams, G., Wong, K.K.,Jr., Podsakoff, G., Forman, S.J. & Chatterjee, S.
  • AAV-CFTR adeno-associated virus
  • Escherichia coli RuvC resolvase The palindromic LTR-LTR junction of Moloney murine leukemia virus is not an efficient substrate for proviral integration. J. Virol. 63, 2629-2637 (1989). McLaughlin, S.K., Collis, P., Hermonat, P.L. & Muzyczka, N.
  • Adeno-associated virus general transduction vectors Analysis of proviral structures. J Virol. 62, 1963-1973 (1988). Miao, CH. et al. The kinetics of rAAV integration in the liver [letter]. Nat Genet 19, 13-15 (1998). Muzyczka, N. Use of adeno-associated virus as a general transduction vector for mammalian cells. Curr. Top. Microbiol. Immunol. 158, 97-129 (1992).
  • Adeno-associated virus type 2-mediated gene transfer correlation of tyrosine phosphorylation of the cellular single-stranded D sequence- binding protein with transgene expression in human cells in vitro and murine tissues in vivo. J Virol 72, 1593-1599 (1998). Qing, K. et al. Human fibroblast growth factor receptor 1 is a co-receptor for infection by adeno-associated virus 2. Nat Med 5, 71-77 (1999). Reich, N.C., Sarnow, P., Duprey, E., and Levine, A.J. (1983). Monoclonal antibodies which recognize native and denatured forms of the adenovirus

Abstract

Cette invention se rapporte à une molécule d'ADN isolée et purifiée comprenant au moins un segment d'ADN, une sous-unité biologiquement active ou variante de cette sous-unité, d'un intermédiaire circulaire de virus adéno-associé, ce segment d'ADN assurant une plus grande stabilité, persistance ou abondance épisomique de la molécule d'ADN isolée dans une cellule hôte. Cette invention concerne également une composition comprenant au moins deux vecteurs de virus adéno-associés.
EP00970689A 1999-10-07 2000-10-06 Virus adeno-associe et utilisations correspondantes Withdrawn EP1224313A1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US15820999P 1999-10-07 1999-10-07
US158209P 1999-10-07
PCT/US2000/027863 WO2001025465A1 (fr) 1999-10-07 2000-10-06 Virus adeno-associe et utilisations correspondantes

Publications (1)

Publication Number Publication Date
EP1224313A1 true EP1224313A1 (fr) 2002-07-24

Family

ID=22567109

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00970689A Withdrawn EP1224313A1 (fr) 1999-10-07 2000-10-06 Virus adeno-associe et utilisations correspondantes

Country Status (4)

Country Link
EP (1) EP1224313A1 (fr)
JP (1) JP2003533170A (fr)
CA (1) CA2386546A1 (fr)
WO (1) WO2001025465A1 (fr)

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6436392B1 (en) 1998-05-20 2002-08-20 University Of Iowa Research Foundation Adeno-associated virus vectors
EP1190249A2 (fr) 1999-06-08 2002-03-27 University Of Iowa Research Foundation Composes et procedes permettant d'ameliorer la transduction de raav
US7241447B1 (en) 1999-10-07 2007-07-10 University Of Iowa Research Foundation Adeno-associated virus vectors and uses thereof
EP1402023A4 (fr) 2001-05-31 2005-06-15 Univ Rockefeller Methode de generation de vecteurs viraux defectifs pour la replication exempts de virus assistant
CA2453405A1 (fr) * 2001-07-13 2003-01-23 University Of Iowa Research Foundation Virus adeno-associes pseudotypes et utilisations de ces derniers
AU2004227358B8 (en) 2003-03-31 2009-12-10 Targeted Genetics Corporation Compounds and methods to enhance rAAV transduction
EP3151866B1 (fr) 2014-06-09 2023-03-08 Voyager Therapeutics, Inc. Capsides chimériques
EP3215191A4 (fr) 2014-11-05 2018-08-01 Voyager Therapeutics, Inc. Polynucléotides codant pour la dopa décarboxylase et destinés au traitement de la maladie de parkinson
JP6863891B2 (ja) 2014-11-14 2021-04-21 ボイジャー セラピューティクス インコーポレイテッドVoyager Therapeutics,Inc. 調節性ポリヌクレオチド
CN114717264A (zh) 2014-11-14 2022-07-08 沃雅戈治疗公司 治疗肌萎缩性侧索硬化(als)的组合物和方法
US11697825B2 (en) 2014-12-12 2023-07-11 Voyager Therapeutics, Inc. Compositions and methods for the production of scAAV
US10983110B2 (en) 2015-12-02 2021-04-20 Voyager Therapeutics, Inc. Assays for the detection of AAV neutralizing antibodies
US11702672B2 (en) 2016-02-08 2023-07-18 University Of Iowa Research Foundation Methods to produce chimeric adeno-associated virus/bocavirus parvovirus
MA43735A (fr) 2016-03-07 2018-11-28 Univ Iowa Res Found Expression médiée par aav utilisant un promoteur et un activateur synthétiques
EP3448874A4 (fr) 2016-04-29 2020-04-22 Voyager Therapeutics, Inc. Compositions pour le traitement de maladies
US11299751B2 (en) 2016-04-29 2022-04-12 Voyager Therapeutics, Inc. Compositions for the treatment of disease
WO2017201258A1 (fr) 2016-05-18 2017-11-23 Voyager Therapeutics, Inc. Compositions et méthodes de traitement de la maladie de huntington
BR112018073384A2 (pt) 2016-05-18 2019-03-06 Voyager Therapeutics, Inc. polinucleotídeos moduladores
EP3506817A4 (fr) 2016-08-30 2020-07-22 The Regents of The University of California Procédés de ciblage et d'administration biomédicaux, et dispositifs et systèmes pour la mise en oeuvre de ceux-ci
WO2018204786A1 (fr) 2017-05-05 2018-11-08 Voyager Therapeutics, Inc. Compositions et méthodes de traitement de la sclérose latérale amyotrophique (sla)
US11752181B2 (en) 2017-05-05 2023-09-12 Voyager Therapeutics, Inc. Compositions and methods of treating Huntington's disease
JOP20190269A1 (ar) 2017-06-15 2019-11-20 Voyager Therapeutics Inc بولي نوكليوتيدات aadc لعلاج مرض باركنسون
JP7229989B2 (ja) 2017-07-17 2023-02-28 ボイジャー セラピューティクス インコーポレイテッド 軌道アレイガイドシステム
MX2020001187A (es) 2017-08-03 2020-10-05 Voyager Therapeutics Inc Composiciones y métodos para la administración de virus adenoasociados.
US20200237799A1 (en) 2017-10-16 2020-07-30 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
WO2019079240A1 (fr) 2017-10-16 2019-04-25 Voyager Therapeutics, Inc. Traitement de la sclérose latérale amyotrophique (sla)
US11660353B2 (en) 2018-04-27 2023-05-30 Decibel Therapeutics, Inc. Compositions and methods for treating sensorineural hearing loss using otoferlin dual vector systems

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5173414A (en) * 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
EP0932418B1 (fr) * 1996-09-06 2007-12-05 The Trustees Of The University Of Pennsylvania Methode de therapie genique basee sur des virus adeno-associes de recombinaison
US6436392B1 (en) * 1998-05-20 2002-08-20 University Of Iowa Research Foundation Adeno-associated virus vectors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
NAKAI H. ET AL: "Increasing the size of rAAV-mediated expression cassettes in vivo by intermolecular joining of two complementary vectors", NATURE BIOTECHNOLOGY, vol. 18, May 2000 (2000-05-01), pages 527 - 532 *
See also references of WO0125465A1 *

Also Published As

Publication number Publication date
WO2001025465A1 (fr) 2001-04-12
WO2001025465A9 (fr) 2002-11-21
JP2003533170A (ja) 2003-11-11
CA2386546A1 (fr) 2001-04-12

Similar Documents

Publication Publication Date Title
US7241447B1 (en) Adeno-associated virus vectors and uses thereof
US7803622B2 (en) Adeno-associated virus vectors
EP1224313A1 (fr) Virus adeno-associe et utilisations correspondantes
EP1849872A1 (fr) Vecteurs de virus adéno associés et leur utilisation
DE69738351T2 (de) Verfaheren zur durch rekombinante adeno-assoziierte virus-gerichtete gentherapie
US20100278791A1 (en) Adeno-associated virus serotype i nucleic acid sequences, vectors and host cells containing same
JP2008506363A (ja) ベクター内非相同末端パリンドローム配列を有するアデノ随伴ウイルスベクター
PT1046711E (pt) Vírus adeno-associado recombinente
JP2001506132A (ja) Aavベクターの産生における使用のためのリコンビナーゼ活性化可能aavパッケージングカセット
WO1996040272A1 (fr) Transduction de myoblastes par vecteurs de virus adeno-associes
US7972856B2 (en) Targeted gene modification by parvoviral vectors
AU2006202785B2 (en) Adeno-associated viruses and uses thereof
EP1222299A1 (fr) Production de virus associes a l'adenovirus (aav) recombinants mettant en oeuvre un adenovirus comprenant des genes rep/cap associes a l'adenovirus
Class et al. Patent application title: Adeno-associated virus vectors Inventors: John F. Engelhardt (Iowa City, IA, US) Dongsheng Duan (Iowa City, IA, US)
WO2000024917A1 (fr) Modification ciblee de genes par vecteurs parvoviraux
JP2003501042A (ja) 非哺乳動物ウイルス由来のキャリアベクターを使用する、組換えウイルスの産生のための組成物および方法
Engelhardt et al. Adeno-associated virus vectors
AU763063B2 (en) AAV transduction of myoblasts
Xing Development of recombinant AAV vectors for lung specific gene therapy
Kroner‐Lux et al. Delivery Systems for Gene Therapy: Adeno‐Associated Virus

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20020507

AK Designated contracting states

Kind code of ref document: A1

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

AX Request for extension of the european patent

Free format text: AL PAYMENT 20020507;LT PAYMENT 20020507;LV PAYMENT 20020507;MK PAYMENT 20020507;RO PAYMENT 20020507;SI PAYMENT 20020507

RIN1 Information on inventor provided before grant (corrected)

Inventor name: DUAN, DONGSHENG

Inventor name: ENGELHARDT, JOHN, F.

Inventor name: YAN, ZIYING

17Q First examination report despatched

Effective date: 20031119

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

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

18D Application deemed to be withdrawn

Effective date: 20090709