WO2012158757A1 - Proviral plasmids for production of recombinant adeno-associated virus - Google Patents

Proviral plasmids for production of recombinant adeno-associated virus Download PDF

Info

Publication number
WO2012158757A1
WO2012158757A1 PCT/US2012/038063 US2012038063W WO2012158757A1 WO 2012158757 A1 WO2012158757 A1 WO 2012158757A1 US 2012038063 W US2012038063 W US 2012038063W WO 2012158757 A1 WO2012158757 A1 WO 2012158757A1
Authority
WO
WIPO (PCT)
Prior art keywords
sequence
plasmid
promoter
gene
itr
Prior art date
Application number
PCT/US2012/038063
Other languages
French (fr)
Inventor
Jeannette BENNICELLI
Jean Bennett
Original Assignee
The Trustees Of The University Of Pennsylvania
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 The Trustees Of The University Of Pennsylvania filed Critical The Trustees Of The University Of Pennsylvania
Priority to US14/117,312 priority Critical patent/US9249425B2/en
Publication of WO2012158757A1 publication Critical patent/WO2012158757A1/en
Priority to US14/979,786 priority patent/US9896665B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • 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
    • 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
    • C12N2750/14121Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • 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
    • 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
    • C12N2750/14151Methods of production or purification of viral material
    • C12N2750/14152Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/60Vector systems having a special element relevant for transcription from viruses
    • 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
    • C12N2830/80Vector systems having a special element relevant for transcription from vertebrates
    • C12N2830/90Vector systems having a special element relevant for transcription from vertebrates avian

Definitions

  • rAAV Recombinant adeno-associated virus
  • ITRs AAV inverted terminal repeats
  • the gene expression cassette for insertion into the rAAV conventionally contains the therapeutic gene and the cis- regulatory elements including a promoter and a polyadenylation signal necessary for gene expression.
  • a gene expression cassette, located between the ITRs is packaged in rAAV particles, which are then used in therapeutic applications.
  • the design and construction of the components, such as the plasmids and gene expression cassettes necessary for producing a recombinant AAV, can be quite labor intensive, due to the variety of plasmids and vectors available, and the need to modify the genes to fit into the appropriate plasmids. This complexity is further increased by pharmaceutical industry and governmental requirements that govern the process for obtaining approval of an rAAV for pharmaceutical use.
  • the invention described herein involves novel AAV proviral vectors characterized by ease of subcloning and which facilitate the ready substitution of alternative plasmid components. These vectors improve the efficiency of rAAV vector production and function.
  • a proviral plasmid is provided in which all the functional elements of the proviral plasmids are modular and readily removable or replaceable by virtue of the unique flanking restriction sites. The modular aspect of these plasmids permits the expression of AAVs with different transgenes.
  • a proviral plasmid comprises a wildtype AAV2 ITR sequence, the ITR flanked by unique restriction sites that permit ready removal from the plasmid or replacement of the entire ITR.
  • the proviral plasmid comprises a 5' ITR and a 3' ITR as described above, each flanked by different unique restriction sites to permit removal or replacement of each ITR individually.
  • a proviral plasmid comprises a modular gene expression cassette comprising in operative association, (i) a wildtype 5' AAV2 ITR sequence flanked by unique restriction sites that permit ready removal or replacement of said ITR;
  • a CMV promoter comprising an upstream non-coding sequence of about 49 nucleotides of CMV to enhance the productivity of the promoter, flanked by unique restriction sites that permit ready removal or replacement of the entire promoter sequence;
  • this plasmid contains a cytomegalovirus (CMV) promoter and further comprises an upstream non-coding sequence of about 49 nucleotides of CMV to enhance the productivity of the promoter.
  • CMV cytomegalovirus
  • this plasmid contains a hybrid promoter comprising a CMV promoter sequence and a chicken beta actin (CBA) promoter sequence and further comprises an upstream non-coding sequence of about 49 nucleotides of CMV to enhance the productivity of the promoter.
  • CBA chicken beta actin
  • the entire promoter is flanked by unique restriction sites that permit ready removal and/or replacement of the enhancer/hybrid promoter sequence.
  • the proviral plasmids described above further comprise a plasmid backbone comprising the elements necessary for replication in bacterial cells, and further comprise a kanamycin resistance gene (Kan R ), the plasmid backbone containing 5' and 3' transcriptional terminator/insulator sequences that isolate transcription in the backbone from transcription in the gene cassette.
  • the proviral plasmids described above further comprise a plasmid backbone comprising the elements necessary for replication in bacterial cells, and further comprise a kanamycin resistance gene ( an R ), the plasmid backbone containing 5' and 3' transcriptional terminator/insulator sequences that isolate transcription in the backbone from transcription in the gene cassette and a lambda stuffer sequence.
  • a proviral plasmid is p604 of FIG.1 SEQ ID NO: 1.
  • a proviral plasmid is p617 of FIG. 2 SEQID NO: 2.
  • a proviral plasmid is p618 of FIG. 3 SEQ ID NO: 3.
  • any of the proviral plasmids described above further comprises a gene encoding sequence inserted into the polylinker sequence.
  • a method for generating a proviral plasmid as described herein.
  • a method for generating a rAAV using any of the proviral plasmids described herein.
  • FIG. 1 is a map showing the features of the 4879 bp AAV proviral plasmid pAAV.CMV.eGFP.wtlTRs (p604).
  • This plasmid contains a modular gene expression cassette in a plasmid backbone based on the pJ201 plasmid (DNA2.0, Menlo Park, CA).
  • the numbers in parentheses are the nucleotide number in SEQ ID NO: 1.
  • the plasmid backbone contains a kanamycin resistance gene (9-803), pTR (1063- 1079), and nrn B1B2T1 txn terminator sequence (988-1162), a pTF3 (3535-3560), a bla txn terminator (3444-3744), an rpn txn terminator (3751-3864), the restriction site Aflll (4057) which may be employed for insertion of a stuffer and a pUC origin of replication (4036-4839).
  • the gene expression cassette comprises in operative association:
  • a cytomegalovirus (CMV) promoter (1443-2018) comprising a 49 nucleotide CMV enhancer sequence extension, the CMV enhancer and minimal CMV promoter, the entire promoter flanked 5' by the unique restriction site Nhel (1428) and 3' by the unique restriction sites BstXI (2202) and Notl (2208), and the 49 nucleotide enhancer sequence flanked in a 239 nucleotide sequence by 5' Nhel (1428) and a 3' unique restriction site Ndel (1667);
  • CMV cytomegalovirus
  • eGFP green fluorescent protein
  • BGH poly A bovine growth hormone polyadenylation signal (2952-3173), flanked 5* by unique restriction sites Hind III (2937), BamHI (2943) and Bglll (2952) and 3' by unique restriction site Xhol (3173) ;
  • FIG. 2 is a map showing the features of the 5060 bp AAV proviral plasmid pAAV.CMV.CBA.synlTR.short (p617).
  • This plasmid contains a modular gene expression cassette in a plasmid backbone based on the pJ201 plasmid (DNA2.0, Menlo Park, CA).
  • the numbers in parentheses are the nucleotide number in SEQ ID NO: 2.
  • the plasmid backbone contains a kanamycin resistance gene (9-803), pTR (1063- 1079), rrn Bl B2 Tl txn terminator sequence (988-1162), a pTF3 (3716-3741), a bla txn terminator (3625-3925), an rpn txn terminator (3932-4045), the restriction site Aflll (4057) which may be employed for insertion of a staffer, and a pUC origin of replication (4217-5020).
  • the gene expression cassette comprises in operative association:
  • a promoter comprising a hybrid CMV/CBA promoter (1443- 3104), derived from the pDRIVE CAG plasmid (Invivogen, San Diego, CA) with an upstream extension of about 49 nucleotides of CMV enhancer; the entire promoter sequence including the upstream sequence is flanked 5' by the unique restriction site Nhel (1428) and 3' by the unique restriction sites Notl (3107), BamHI (3124), Pstl (3133) and Bglll (3133); the enhancer sequence extension is contained within a 239 nucleotide sequence as described for p604;
  • BGH poly A bovine growth hormone polyadenylation signal (3133-3354), flanked 5' by unique restriction sites Notl (3107), BamHI (3124), Pstl (3133) and Bglll (3133); and 3' by unique restriction site Xhol (3354);
  • FIG. 3 is a map showing the features of the 10131 bp AAV pro viral plasmid pAAV.CMV.CBA.synlTR.long.stuffer 15 or p618 (SEQ ID NO: 3).
  • the numbers in parentheses are the nucleotide numbers in SEQ ID NO: 3.
  • This plasmid contains a modular gene expression cassette in a plasmid backbone based on the pJ201 plasmid (DNA2.0, Menlo Park, CA).
  • the plasmid backbone contains a kanamycin resistance gene (9-803), pTR (1063-1079), rrn Bl B2 Tl txn terminator sequence (988- 1 162), a pTF3 (3716-3741), a bla txn terminator (3625-3925), an rpn txn terminator (3932- 4045), a lambda stuffer sequence (4061-9127) which is inserted into the site Aflll, and a pUC origin of replication (9288-10091).
  • the gene expression cassette comprises in operative association:
  • a promoter comprising a hybrid CMV/CBA promoter, derived from the pDRIVE CAG plasmid (Invivogen, San Diego, CA) with an upstream extension of about 49 nucleotides of CMV enhancer, the entire promoter (1443-3104) flanked 5' by the unique restriction site Nhel (1428) and 3' by the unique restriction sites Notl (3107), Pstl (3133) and Bglll (3133); the enhancer sequence extension is contained within a 239 nucleotide sequence as described for p604;
  • BGH poly A bovine growth hormone polyadenylation signal
  • FIG. 4 is a cartoon of the synthesized Exon 1 ORF of hRdCVFl (DNA2.0) and the modifications made to it for insertion into a proviral plasmid. The modifications include the addition of Notl and BamHI restriction sites, a stop codon embedded in Bell site to facilitate addition of epitope tag, and a complete Kozak consensus sequence that overlaps Notl. Only the hRdCVFl ORF of SEQ ID NO:4 is included as the transgene. The entire sequence depicts 357 bp.
  • FIG. 5 is a cartoon of a genomic fragment of hRdCVFl containing the Exon 1 ORF, exonic sequence upstream of the start codon, and intronic sequence downstream of stop codon. Modifications incorporated during synthesis (DNA2.0) include the addition of Notl and BamHI restriction sites. The entire sequence depicts 470bp.
  • FIG. 6 is a gel showing the results of a Western blot analysis of hRdCVFl expression in cultured cells after transfection, as described in Example 2.
  • FIG. 7 is a sequence showing the alignment of hRdCVFl genomic fragment of 456 bases (top line, SEQ ID NO: 4) and open reading frame or exon 1 ORF of 327 bases (bottom line; SEQ ID NO: 5).
  • novel AAV proviral plasmids described herein can be used to increase the productivity and efficiency of generating a variety of rAAV expressing a variety of targeted genes. All the functional elements of the proviral plasmids are modular and readily removable or replaceable by virtue of the unique flanking restriction sites. The modular aspect of these plasmids permits the efficient generation of AAVs which express different genes.
  • a proviral plasmid is designed to include a plasmid
  • backbone and a “minigene” or “gene expression cassette” flanked by AAV ITRs.
  • ITRs and minigene comprise the recombinant AAV (rAAV) genome that is packaged into AAV particles.
  • the gene expression cassette includes, at a minimum, a transgene (i.e., the gene desired to be transported by the rAAV and expressed in selected cells) and its regulatory sequences, flanked by 5 ' and 3 ' AAV inverted terminal repeats (ITRs).
  • a transgene i.e., the gene desired to be transported by the rAAV and expressed in selected cells
  • ITRs 5 ' and 3 ' AAV inverted terminal repeats
  • Every significant component in the rAAV genome contained within the proviral plasmid is flanked by unique restriction sites, i.e., restriction sites used only once in the plasmids to enable ready removal and/or replacement of individual components in the rAAV genome.
  • unique restriction site is meant a restriction site that is cleaved by an enzyme that cannot cleave another restriction site in the proviral plasmid.
  • the unique flanking restriction enzyme sites allow directional cloning of one or more components into the plasmid.
  • a pair of unique restriction enzyme sites flanking one component allows digestion at only a single locus in the proviral plasmid and ready insertion or deletion of only one component of the recombinant AAV genome.
  • suitable restriction enzymes for unique use in the desired plasmids include those identified in the description of the figures and in the figures themselves. Suitable restriction enzymes may be identified using information readily available to those of skill in the art in the literature and in a variety of on-line databases, e.g., the REBASETM database. Suitable restriction enzymes for the use in generating a proviral plasmid of this invention can be readily determined using a variety of computer programs and/or on-line databases. Suitable restriction enzymes are available from a variety of commercial sources including, e.g., New England Biolabs, Obiogene, Lift Technology, Roche, Clontech, Stratagene, Amersham, Pharmacia, among others.
  • a proviral plasmid contains a wildtype ITR of AAV serotype 2.
  • the ITRs are synthesized to remove a common mutation, i.e., a deletion in the upstream ITR that is present in many known vectors. This attribute alone allows use of these plasmids to improve packaging efficiency of rAAV during production.
  • the inventors theorized that mutated ITR sequences are corrected in the cell during production of the rAAV.
  • use of the wildtype ITR, particularly the 5' ITR conserves the cell's resources and is more efficient.
  • ITR sequences from other suitable AAV serotype sources may be selected or ITR mutations may be introduced by design to alter function of the rAAV.
  • a significant attribute of these plasmids is that the intact wildtype ITR sequence is flanked by unique restriction sites that permit ready removal or replacement of the ITR from the plasmid.
  • the proviral plasmid comprises a 5' ITR flanked by unique restriction sites to permit its removal or replacement.
  • the proviral plasmid comprises a 3' ITR flanked by unique restriction sites to permit its removal or replacement.
  • the proviral plasmid contains a 5' ITR and a 3' ITR, each ITR flanked by unique restriction sites, so that each ITR may be individually removed from the plasmid, if desired.
  • the proviral plasmids described in the FIGs. 1 -3 have identified exemplary ITRs.
  • This modular capacity of the ITRs in the proviral plasmids allows for ready insertion and replacement of a variety of ITR sequences and permits use of the proviral plasmids in a number of methods for generating recombinant viruses.
  • proviral plasmids also include regulatory elements which are operably linked to the transgene in a manner which permits the transgene's transcription, translation and/or expression in a cell transfected with the plasmid vector or infected with the virus produced generated by use of the proviral plasmids.
  • operably linked sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest.
  • Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation (polyA) signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e. , Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance secretion of the encoded product.
  • polyA polyadenylation
  • a great number of expression control sequences, including promoters which are native, constitutive, inducible and/or tissue-specific, are known in the art and may be utilized.
  • proviral plasmids are designed so that the promoter and poly A sequences are modular and may be readily replaced with other known promoter and polyA sequences.
  • the promoter is a cytomegalovirus promoter (p604) comprising an enhancer and minimal promoter with a 49 nucleotide upstream extension of the enhancer sequence
  • the promoter is a hybrid promoter comprising a CMV enhancer sequence and a chicken beta actin (CBA) promoter sequence (the pDRIVE-CAG promoter (Invitrogen).
  • CBA chicken beta actin
  • the hybrid CMV/CBA promoter further comprises the upstream non-coding sequence of about 49 nucleotides of CMV SEQ ID NO: 6 to enhance the productivity of the promoter, wherein the upstream CMV enhancer/extension sequence is in the larger 239 sequence, discussed above is flanked by unique restriction sites that permit ready removal or replacement of the upstream CMV sequence from the hybrid promoter sequence.
  • these promoter sequences are identified.
  • the CMV/CBA hybrid promoter (Invivogen) was about twice as strong a promoter when it is preceded by about a sequence of about 49 nucleotides upstream of the CMV promoter sequence.
  • this upstream enhancer sequence is flanked by unique restriction sites, permitting its removal and/or replacement should the strong promoter be too strong in expressing any particular transgene in the selected host cell.
  • the 49 nucleotide extension SEQ ID NO: 6 is flanked 5' by unique restriction site Nhel and further downstream to create the 239 nucleotide sequence by unique restriction site Ndel.
  • the proviral plasmids allow for the easy removal or replacement of the upstream CMV enhancer sequence alone by cleavage at the unique restriction sites flanking the enhancer sequence.
  • the proviral plasmids permit removal or replacement of the entire promoter sequence
  • promoter/enhancer sequence by the presence of unique restriction sites at both ends of the entire promoter sequence.
  • these proviral plasmids allow for excision of the existing promoters and insertion between the unique sites of any number of conventionally employed promoters.
  • Suitable constitutive or inducible promoter sequences are available from a variety of commercial sources, including, without limitation, Invitrogen, Clontech and Ariad. Many other systems, e.g., native promoters, tissue-specific promoters, etc., have been described and can be readily selected by one of skill in the art. See, e.g. , the promoters identified in US Patent No. 7,906, 111, incorporated by reference herein.
  • Another modular regulatory sequence in the proviral plasmids described herein is the polyadenylation sequence.
  • the bovine growth hormone polyA site is flanked by unique restriction sites, thereby permitting its ready replacement or removal, as desired.
  • the selection of the particular polyA sequence is not critical. It is a characteristic of these plasmids that quick replacement of this regulatory sequence is possible depending upon the selection of transgene, promoter and/or host cell.
  • the proviral plasmids described herein also contain a multiple cloning site/polylinker sequence for ready insertion of almost any transgene without the requirement for extensive modification of the transgene. In one embodiment this is due to the inclusion of a number of unique restriction sites, such as Notl, BamHI, Pstl and Bglll.
  • the polylinker sequences are located between the promoter sequence and the polyA sequence. Once a transgene is selected, the transgene (or a portion thereof, if it is a large transgene), may be inserted into the polylinker site of these plasmids.
  • the transgene is a nucleic acid sequence, heterologous to the ITR and regulatory sequences flanking the transgene, which encodes a polypeptide, protein, RNA, or other product of interest.
  • the nucleic acid coding sequence is operatively linked to regulatory components in a manner which permits transgene transcription, translation, and/or expression in a host cell.
  • transgene sequence will depend upon the use to which the resulting vector will be put.
  • one type of transgene sequence includes a reporter sequence, which upon expression produces a detectable signal.
  • a reporter sequence is enhanced GFP (eGFP).
  • eGFP enhanced GFP
  • the eGFP gene may be easily removed and replaced by any gene up to about 3.3 kb in length for expression in rAAV using current packaging protocols.
  • such reporter sequences include, without limitation, DNA sequences encoding ⁇ -lactamase, ⁇ - galactosidase (LacZ), alkaline phosphatase, thymidine kinase, green fluorescent protein (GFP), chloramphenicol acetyltransferase (CAT), luciferase, membrane bound proteins including, for example, CD2, CD4, CD8, the influenza hemagglutinin protein, and others well known in the art, to which high affinity antibodies directed thereto exist or can be produced by conventional means, and fusion proteins comprising a membrane bound protein appropriately fused to an antigen tag domain from, among others, hemagglutinin or Myc.
  • DNA sequences encoding ⁇ -lactamase, ⁇ - galactosidase (LacZ), alkaline phosphatase, thymidine kinase, green fluorescent protein (GFP), chloramphenicol acetyltransferase (CAT), luci
  • coding sequences when associated with regulatory elements which drive their expression, provide signals detectable by conventional means, including enzymatic, radiographic, colorimetric, fluorescence or other spectrographic assays, fluorescent activating cell sorting assays and immunological assays, including enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA) and immunohistochemistry.
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • immunohistochemistry immunohistochemistry.
  • the marker sequence is the LacZ gene
  • the presence of the vector carrying the signal is detected by assays for beta-galactosidase activity.
  • the transgene is green fluorescent protein or luciferase
  • the vector carrying the signal may be measured visually by color or light production in a luminometer.
  • the transgene is a non-marker sequence encoding a product which is useful in biology and medicine, such as proteins, peptides, RNA, enzymes, dominant negative mutants, or catalytic RNAs.
  • Desirable RNA molecules include tRNA, dsRNA, ribosomal RNA, catalytic RNAs, siRNA, small hairpin RNA, trans-splicing RNA, and antisense RNAs.
  • a useful RNA sequence is a sequence which inhibits or extinguishes expression of a targeted nucleic acid sequence in the treated animal.
  • suitable target sequences include oncologic targets and viral diseases. See, for examples of such targets the oncologic targets and viruses identified in US Patent No.
  • the transgene is the human rod derived cone variability factor 1 (see e.g., NCBI Accession No. NT_01 1295.11 and NM_138454.1, incorporated by reference; SEQ ID NO: 4).
  • the transgene is exon 1 open reading frame (ORF) of the hRdCVFl cDNA, e.g., from the Ncol site at nucleotide 12 to the stop codon and flanked by Notl and BamHI restriction sites (SEQ ID NO: 5; FIG 4).
  • the transgene is the exon 1 ORF with exonic sequences upstream of the start codon and intronic sequences downstream of the stop codon flanked by the insertion of a Notl restriction site at nucleotide 3 of the hRdCVf exon 1 fragment and a BamHI restriction site at nucleotide position 466 of the fragment (FIG. 5).
  • the hRdCVf 1 exon 1 ORF is modified by embedding the stop codon in a Bell site at nucleotide position 341 to facilitate the addition of an epitope tag (FIG. 4). A complete Kozak consensus sequence was also included and overlaps the Notl sequence (FIG. 4).
  • the transgene may be used to correct or ameliorate gene deficiencies, which may include deficiencies in which normal genes are expressed at less than normal levels or deficiencies in which the functional gene product is not expressed.
  • the transgene may provide a product to a cell which is not natively expressed in the cell type or in the host.
  • a preferred type of transgene sequence encodes a therapeutic protein or polypeptide which is expressed in a host cell.
  • the invention further includes using multiple transgenes. In certain situations, a different transgene may be used to encode each subunit of a protein, or to encode different peptides or proteins or portions of a single large protein.
  • the size of the DNA encoding the protein subunit is large, e.g., for an immunoglobulin, the platelet-derived growth factor, or a dystrophin protein. Additionally this may be desirable for proteins encoded by genes that are mutated in inherited forms of blindness such as centrosomal protein 290kDa (CEP290) mutations in Leber Congenital Amaurosis and ATP-binding cassette, sub-family A member 4 (ABCR).mutations in Stargardt's Disease.
  • CEP290 centrosomal protein 290kDa
  • ABCR sub-family A member 4
  • a single transgene includes the DNA encoding each of the subunits, with the DNA for each subunit separated by an internal ribo2yme entry site (IRES).
  • IRES internal ribo2yme entry site
  • the DNA may be separated by sequences encoding a 2A peptide, which self-cleaves in a post- translational event. See, e.g., M.L. Donnelly, et al, J. Gen.
  • a first AAV may carry an expression cassette which expresses a single transgene and a second AAV may carry an expression cassette which expresses a different transgene for co- expression in the host cell.
  • the selected transgene may encode any biologically active product or other product, e.g., a product desirable for study.
  • transgenes may be readily selected by one of skill in the art.
  • the selection of the transgene is not considered to be a limitation of this invention, but may be selected from among many known transgenes (see, e.g., transgenes identified in US Patent No. 7,906,111, incorporated by reference herein).
  • plasmid p604 can incorporate transgenes of up to 3.3 kb in length for expression in rAAV; and p617 and p618 can incorporate genes up to 2.4 kb in length.
  • the plasmid backbone of the proviral plasmid includes the conventional elements necessary for replication and, optionally, integration in prokaryotic cells (e.g., bacterial cells).
  • the plasmid backbone is pJ201 (DNA2.0). See FIGs. 1-3. Still other backbones may be employed if desired expression is mammalian cells, or both.
  • the backbone desirably contains a selectable marker, e.g., a kanamycin resistance gene (Kan R ).
  • the selectable marker gene is located in the backbone of the proviral plasmid so that it will not be rescued and incorporated into the resulting recombinant virus when the proviral plasmid is used to produce rAAV. It can be used to signal the presence of the plasmids in bacterial cells. Kan R is particularly useful in these proviral plasmids for the generation of rAAV which are clinically useful or for generation of rAAV used in the treatment of humans for diseases or disorders requiring rAAV- mediated delivery of a particular transgene.
  • a selectable marker e.g., a kanamycin resistance gene (Kan R ).
  • Kan R is particularly useful in these proviral plasmids for the generation of rAAV which are clinically useful or for generation of rAAV used in the treatment of humans for diseases or disorders
  • another optional component of the plasmid backbone is an origin of replication, such as the pUC origin of replication identified in FIGs. 1-3.
  • the plasmid backbone contains 5' and 3' transcriptional terminator/insulator sequences that isolate transcription in the backbone from transcription in the gene cassette.
  • sequences exemplified in the FIGs. 1 -3 are pTR, nrn B1B2T1 txn terminator sequence, pTF3, a bla txn terminator, and an rpn txn terminator.
  • the plasmid backbone further comprises a stuffer sequence, such as the stuffer sequence exemplified in p618 (see, FIGs. 2 and 3).
  • a stuffer sequence is a non-coding sequence used to enlarge the backbone.
  • the stuffer sequence contains no functional elements from its viral origin. It does not transcribe genes and contains no promoter.
  • the stuffer in the case of p618, is a 5.1 kb sequence derived from lambda phage.
  • the stuffer increases the size of the proviral plasmid to greater than 8 kb. It prevents the backbone ⁇ i.e., all sequence outside of the recombinant AAV genome) from being packaged into the rAAV capsid, which has a capacity of only 4.7 kb.
  • proviral plasmids Given this description of the proviral plasmids herein, one of skill in the art is expected to be able to select these and other plasmid backbone elements. Many such sequences are available (see, e.g., Sambrook et al, and references cited therein).
  • the modular proviral plasmids p604, p617 and p618, and variants thereof containing selected transgenes desirably incorporate a variety of the components described herein.
  • p618 in particular is designed as a plasmid that can be used to make a vector for a clinical trial. These features include all of the necessary elements for expression and packaging, except the gene of interest. Any gene can be easily cloned into the multiple cloning site for cDNA that includes Notl, BamHI, Pstl, and BgHI.
  • proviral plasmids as described herein improves the ease of subcloning transgenes of a variety of types and sizes into recombinant AAV genomes, facilitates the substitution of alternative plasmid features, and improves the efficiency of rAAV vector production and function.
  • These plasmids may be made by resort to conventional techniques given the descriptions, including sequences provided herein. The method of making the proviral plasmids given this disclosure is within the skill of the art.
  • the proviral plasmids may be employed in currently conventional packaging methodologies to generate a recombinant virus expressing the transgene carried by the proviral plasmids.
  • Suitable production cell lines are readily selected by one of skill in the art.
  • a suitable host cell can be selected from any biological organism, including prokaryotic ⁇ e.g., bacterial) cells, and eukaryotic cells, including, insect cells, yeast cells and mammalian cells.
  • the proviral plasmid is transfected into a selected packaging cell, where it may exist transiently.
  • the minigene or gene expression cassette with its flanking ITRs is stably integrated into the genome of the host cell, either chromosomally or as an episome.
  • Suitable transfection techniques are known and may readily be utilized to deliver the recombinant AAV genome to the host cell.
  • the proviral plasmids are cultured in the host cells which express the cap and/or rep proteins.
  • the gene expression cassettes with flanking AAV ITRs are rescued and packaged into the capsid protein or envelope protein to form an infectious viral particle.
  • the proviral plasmid when delivering the vector comprising the minigene by transfection, is delivered in an amount from about 5 ⁇ g to about 100 ⁇ g DNA, about 10 ⁇ g to about 50 g DNA to about 1 x 10 4 cells to about 1 x 10 13 cells, or about 1 x 10 5 cells.
  • the relative amounts of plasmid DNA to host cells may be adjusted, taking into consideration such factors as the selected proviral plasmid, the delivery method and the host cells selected.
  • adeno-associated virus (AAV) containing a transgene carried by the p618 plasmid
  • AAV adeno-associated virus
  • the p618 plasmid is co-transfected into adenovirus-infected human embryonic kidney 293 (293) cells with a plasmid providing the AAV helper functions.
  • 293 cells are transfected with three plasmids: p618 containing a transgene, a plasmid providing the AAV helper function, and a third plasmid that substitutes for the wild type (wt) adenovirus by providing E2A, E4 and VA adenoviral genes to enable viral replication.
  • the second method offers the advantage of avoiding wt adenovirus infection and of yielding rAAV preparations that are presumed to be free of adenovirus. See, Shi et al, Virology J., 6:3 (January 2009).
  • the components required to be cultured in the host cell to package a recombinant AAV genome in an AAV capsid may be provided to the host cell in trans.
  • any one or more of the required components e.g., recombinant AAV genome, rep sequences, cap sequences, and/or helper functions
  • a stable host cell which has been engineered to contain one or more of the required components using methods known to those of skill in the art.
  • a stable host cell will contain the required component(s) under the control of an inducible promoter.
  • the required component(s) may be under the control of a constitutive promoter. Examples of suitable inducible and constitutive promoters are provided herein, in the discussion of regulatory elements suitable for use with the transgene.
  • a selected stable host cell may contain selected component(s) under the control of a constitutive promoter and other selected component(s) under the control of one or more inducible promoters.
  • a stable host cell may be generated which is derived from 293 cells (which contain El helper functions under the control of a constitutive promoter), but which contains the rep and/or cap proteins under the control of inducible promoters.
  • Still other stable host cells may be generated by one of skill in the art.
  • the rep sequences, cap sequences, and helper functions required for producing the rAAV of the invention may be delivered to the packaging host cell in the form of any genetic element which transfer the sequences carried thereon.
  • the selected genetic element may be delivered by any suitable method, including those described herein.
  • any embodiment of this invention are known to those with skill in nucleic acid manipulation and include genetic engineering, recombinant engineering, and synthetic techniques.
  • Such techniques include conventional cloning techniques of cDNA such as those described in texts (Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, NY.), use of overlapping oligonucleotide sequences of the virus genomes, polymerase chain reaction, and any suitable method which provides the desired nucleotide sequence.
  • Standard transfection and co-transfection techniques are employed, e.g. , CaP0 4 precipitation techniques.
  • the AAV and components described herein may be isolated or obtained from academic, commercial, or public sources (e.g., the American Type Culture Collection, Manassas, VA). Alternatively, the AAV sequences may be obtained through synthetic or other suitable means by reference to published sequences such as are available in the literature or in databases such as, e.g., GenBank®, PubMed®, or the like.
  • the host cell contains the sequences which drive expression of a novel AAV capsid protein of the invention (or a capsid protein comprising a fragment thereof) in the host cell and rep sequences of the same source as the source of the AAV ITRs found in the recombinant AAV genome, or a cross-complementing source.
  • the AAV cap and rep sequences may be independently obtained from an AAV source as described above and may be introduced into the host cell in any manner known to one in the art as described above.
  • the sequences encoding each of the essential rep proteins may be supplied by different AAV sources ⁇ e.g., AAVl , AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8).
  • the rep78/68 sequences may be from AAV2
  • the rep52/40 sequences may be from AAV8.
  • the host cell stably contains the capsid protein under the control of a suitable promoter, such as those described above.
  • the capsid protein is expressed under the control of an inducible promoter.
  • the capsid protein is supplied to the host cell in trans.
  • the capsid protein may be delivered via a plasmid which contains the sequences necessary to direct expression of the selected capsid protein in the host cell.
  • the plasmid carrying the capsid protein also carries other sequences required for packaging the rAAV, e.g., the rep sequences.
  • the host cell stably contains the rep sequences under the control of a suitable promoter, such as those described above.
  • the essential rep proteins are expressed under the control of an inducible promoter.
  • the rep proteins are supplied to the host cell in trans.
  • the rep proteins may be delivered via a plasmid which contains the sequences necessary to direct expression of the selected rep proteins in the host cell.
  • the plasmid carrying the capsid protein also carries other sequences required for packaging the rAAV, e.g., the rep and cap sequences.
  • the rep and cap sequences may be transfected into the host cell on a single nucleic acid molecule and exist stably in the cell as an episome.
  • the rep and cap sequences are stably integrated into the chromosome of the cell.
  • Another embodiment has the rep and cap sequences transiently expressed in the host cell.
  • a useful nucleic acid molecule for such transfection comprises, from 5' to 3', a promoter, an optional spacer interposed between the promoter and the start site of the rep gene sequence, an AAV rep gene sequence, and an AAV cap gene sequence.
  • the rep and/or cap sequences may be supplied on a vector that contains other DNA sequences that are to be introduced into the host cells.
  • the vector may contain the rAAV construct comprising the minigene.
  • the vector may comprise one or more of the genes encoding the helper functions, e.g., the adenoviral proteins El , E2a, and E4 ORF6, and the gene for VAI RNA.
  • the promoter used in this construct may be any of the constitutive, inducible or native promoters known to one of skill in the art or as discussed above.
  • the selection of the AAV to provide any of these sequences does not limit the invention.
  • the promoter for rep is an inducible promoter, such as are discussed above in connection with the transgene regulatory elements.
  • One preferred promoter for rep expression is the T7 promoter.
  • the vector comprising the rep gene regulated by the T7 promoter and the cap gene, is transfected or transformed into a cell which either constitutively or inducibly expresses the T7 polymerase. See
  • the spacer is an optional element in the design of the vector.
  • the spacer is a DNA sequence interposed between the promoter and the rep gene ATG start site.
  • the spacer may have any desired design; that is, it may be a random sequence of nucleotides, or alternatively, it may encode a gene product, such as a marker gene.
  • the spacer may contain genes which typically incorporate start/stop and polyA sites.
  • the spacer may be a non-coding DNA sequence from a prokaryote or eukaryote, a repetitive non-coding sequence, a coding sequence without transcriptional controls or a coding sequence with transcriptional controls.
  • Two exemplary sources of spacer sequences are the phage ladder sequences or yeast ladder sequences, which are available commercially, e.g.
  • the spacer may be of any size sufficient to reduce expression of the rep78 and rep6E gene products, leaving the rep52, repAO and cap gene products expressed at normal levels.
  • the length of the spacer may therefore range from about 10 bp to about 10.0 kbp, preferably in the range of about 100 bp to about 8.0 kbp.
  • the spacer is preferably less than 2 kbp in length; however, the invention is not so limited.
  • the molecule(s) providing rep and cap may exist in the host cell transiently (i.e., through transfection), it is preferred that one or both of the rep and cap proteins and the promoter(s) controlling their expression be stably expressed in the host cell, e.g., as an episome or by integration into the chromosome of the host cell.
  • the methods employed for constructing embodiments of this invention are conventional genetic engineering or recombinant engineering techniques such as those described in the references above.
  • the rep or cap protein may be provided stably by a host cell.
  • the packaging host cell also requires helper functions in order to package the rAAV of the invention.
  • these functions may be supplied by a herpesvirus.
  • the necessary helper functions are each provided from a human or non- human primate adenovirus source, such as those described above and/or are available from a variety of sources, including the American Type Culture Collection (ATCC), Manassas, VA (US).
  • ATCC American Type Culture Collection
  • VA Manassas
  • the host cell is provided with and/or contains an Ela gene product, an Elb gene product, an E2a gene product, and/or an E4 ORF6 gene product.
  • the host cell may contain other adenoviral genes such as VAI RNA, but these genes are not required. In a preferred embodiment, no other adenovirus genes or gene functions are present in the host cell.
  • adenoviral DNA which expresses the Ela gene product it is meant any adenovirus sequence encoding Ela or any functional Ela portion.
  • Adenoviral DNA which expresses the E2a gene product and adenoviral DNA which expresses the E4 ORF6 gene products are defined similarly.
  • any alleles or other modifications of the adenoviral gene or functional portion thereof Such modifications may be deliberately introduced by resort to conventional genetic engineering or mutagenic techniques to enhance the adenoviral function in some manner, as well as naturally occurring allelic variants thereof. Such modifications and methods for manipulating DNA to achieve these adenovirus gene functions are known to those of skill in the art.
  • the adenovirus Ela, Elb, E2a, and/or E40RF6 gene products, as well as any other desired helper functions, can be provided using any means that allows their expression in a cell.
  • Each of the sequences encoding these products may be on a separate vector, or one or more genes may be on the same vector.
  • the vector may be any vector known in the art or disclosed above, including plasmids, cosmids and viruses.
  • adenoviral genes may be stably integrated into the genome of the host cell, stably expressed as episomes, or expressed transiently.
  • the gene products may all be expressed transiently, on an episome or stably integrated, or some of the gene products may be expressed stably while others are expressed transiently.
  • the promoters for each of the adenoviral genes may be selected independently from a constitutive promoter, an inducible promoter or a native adenoviral promoter.
  • the promoters may be regulated by a specific physiological state of the organism or cell (i.e., by the differentiation state or in replicating or quiescent cells) or by exogenously added factors, for example.
  • the host cell itself may be selected from any biological organism, including prokaryotic (e.g., bacterial) cells, and eukaryotic cells, including, insect cells, yeast cells and mammalian cells.
  • Particularly desirable host cells are selected from among any mammalian species, including, without limitation, cells such as A549, WEHI, 3T3, 10T1/2, BH , MDC , COS 1, COS 7, BSC 1, BSC 40, BMT 10, VERO, WI38, HeLa, 293 cells (which express functional adenoviral El), Saos, C2C12, L cells, HT1080, HepG2 and primary fibroblast, hepatocyte and myoblast cells derived from mammals including human, monkey, mouse, rat, rabbit, and hamster.
  • cells such as A549, WEHI, 3T3, 10T1/2, BH , MDC , COS 1, COS 7, BSC 1, BSC 40, BMT 10, VERO, WI38, HeLa, 293 cells (which express functional adenoviral El), Saos, C2C12, L cells, HT1080, HepG2 and primary fibroblast, hepatocyte and myoblast cells derived from mammals including human, monkey
  • the selection of the mammalian species providing the cells is not a limitation of this invention; nor is the type of mammalian cell, i.e., fibroblast, hepatocyte, tumor cell, etc.
  • the requirements for the cell used is that it not carry any adenovirus gene other than El , E2a and/or E4 ORF6; it not contain any other virus gene which could result in homologous recombination of a contaminating virus during the production of rAAV; and it is capable of infection or transfection of DNA and expression of the transfected DNA.
  • the host cell is one that has rep and cap stably transfected in the cell.
  • One host cell useful in the present invention is a host cell stably transformed with the sequences encoding rep and cap, and which is transfected with the adenovirus El , E2a, and E40RF6 DNA and a proviral plasmid carrying the recombinant AAV genome as described above.
  • Stable rep and/or cap expressing cell lines such as B-50 (International Patent Application Publication No. WO 99/15685), or those described in US Patent No. 5,658,785, may also be similarly employed.
  • Another desirable host cell contains the minimum adenoviral DNA which is sufficient to express E4 ORF6.
  • Yet other cell lines can be constructed using the novel AAV9 cap sequences of the invention.
  • the preparation of a host cell according to this invention involves techniques such as assembly of selected DNA sequences. This assembly may be accomplished utilizing conventional techniques. Such techniques include cDNA and genomic cloning, which are well known and are described in Sambrook et al., cited above, use of overlapping oligonucleotide sequences of the adenovirus and AAV genomes, combined with polymerase chain reaction, synthetic methods, and any other suitable methods which provide the desired nucleotide sequence.
  • proviral plasmids discussed herein and the other necessary elements into the host cell may also be accomplished using techniques known to the skilled artisan and as discussed throughout the specification. See, also, S. Shi et al, Virol. J., 6:3 (2009).
  • standard transfection techniques are used, e.g., CaP0 4 transfection or electroporation, and/or infection by hybrid adenoviras/AAV vectors into cell lines such as the human embryonic kidney cell line HEK 293 (a human kidney cell line containing functional adenovirus El genes which provides trans-acting El proteins).
  • compositions of the invention may also be used for production of a desired gene product in vitro.
  • a desired product ⁇ e.g., a protein
  • a desired product may be obtained from a desired culture following transfection of host cells with proviral plasmid containing the desired transgene.
  • the expressed product may then be purified and isolated, as desired. Suitable techniques for transfection, cell culturing, purification, and isolation are known to those of skill in the art.
  • EXAMPLE 1 Construction of AA V proviral plasmids containing human RdCVFl
  • NCBI reference NT_011295.11, Homo sapiens chromosome 19 genomic contig, GRCh37.p2 reference primary assembly contains the nucleotedoxin-like protein 1 or rod- derived cone viability factor 1 (RdCVFl).
  • the mRNA reference sequence is
  • NM_138454.1 (SEQ ID NO: 4).
  • Exon 1 begins at nt 21 of the genomic fragment.
  • two DNA fragments comprising the open reading frame (ORF) and a genomic fragment are synthesized by DNA2.0.
  • the latter is synthesized as a 470 bp fragment with 5' Notl and 3' BamHI sites to facilitate cloning.
  • the 357 bp ORF cDNA is synthesized with a complete ozak consensus sequence that was partly embedded in a 5' Notl site.
  • the stop codon is embedded in a Bell site, followed by a BamHI site.
  • the Bell site is inserted to allow addition of epitope tags.
  • Each fragment was synthesized by DNA2.0 and supplied in a plasmid vector pJ201.
  • Each fragment (ORF and genomic fragment, SEQ ID NOs: 5 and 4, respectively) is subcloned into AAV proviral plasmids pAAV.CMV.CBA.synITR.short (p617), and pAAV.CMV.CBA.synITR.long (p618) and into an additional known plasmid.
  • the resulting plasmids p623/ORF and p624/genomic fragment were derived from p617.
  • the resulting plasmids p625/ORF and p626/genomic fragment are derived from plasmid p618.
  • Plasmids included the newly made AAV-hRdCVFl proviral plasmids described in Example 1, as well as pAAV2.1 CMV.hRdCVFl (p368), another plasmid made by the inventors, and two pcDNA3-based plasmids p452 and p488 supplied by a third party.
  • a Western blot experiment was performed as follows: 293T cells (passage number n+ 5) were seeded into 100 mm culture dishes. At 24 hours after seeding, the cells were approximately 30% confluent and were transfected with 25 ⁇ g/dish of endotoxin-free plasmid (Quiagen) using lipofectamine LTX (ratio 2.5 ⁇ lipofectamine LTX per ⁇ g of plasmid).
  • the cell culture medium was DMEM with high glucose, no pyruvate, and 10% FBS (no antibiotic). At 6 h the medium was aspirated and replaced with 5 ml of serum- free Optimem medium per dish. 24 h post transfection, the conditioned media were collected and spun for 5 min at 1200 rpm to remove cells and debris. A protease inhibitor cocktail was added to the clarified media and they were stored at -80°C until use. Cells were scraped from each dish into PBS, and the collected cells were pooled with the cells collected from the conditioned medium. Each cell pellet was resuspended in 250 ⁇ of RIPA containing protease inhibitor cocktail.
  • conditioned media were thawed and concentrated at 4°C using Amicon Ultra- 15 concentrators with a 3 Kd cut-off. 5 ml of conditioned medium was concentrated to 400 ⁇ volume (12.5x). Samples consisted of 50 ⁇ g cell lysate (in a volume of 21 ⁇ ) or 21 ⁇ of concentrated, conditioned medium. Each sample represented roughly l/20 th of the entire contents of the culture dish. Samples were denatured in lx LDS loading buffer (Invitrogen) with DTT for 10 min at 70°C and loaded onto NuPAGE 4-12% gradient Bis-Tris gels that were subsequently run in MES buffer (Invitrogen). Gels were electroblotted onto Hybond ECL nitrocellulose (Amersham) and the transfer was verified by Ponceau staining.
  • plasmids p624 (lane 5) and p626 (lane 7) express a band just below the 17 kd molecular weight marker that most likely represents a product resulting from an incomplete translational stop at the predicted stop codon immediately after the ORF.
  • plasmids p623, p624 (based on p617), p625 and p626 (based on p618) show increased expression of the 12.7 kd band relative to p621 and p622, indicating that the addition of upstream CMV sequences in the plasmids results in a more powerful promoter.
  • p625 and p626 The best prospects for in vivo use are p625 and p626.
  • the former is best for robust expression of a product of uniform length. The latter would be useful if the longer product is biologically relevant.
  • These two p618-based plasmids are preferred over p623 and p624 that perform similarly, but do not contain a staffer in the plasmid backbone. These plasmids do not contain a deletion in the upstream ITR, and as judged by band intensities in the Western blots, the promoter appears to be twice as strong as the promoter in p621 and p622 which lacks the 49 nucleotide-long extension of the CMV enhancer.
  • human Rod-derived Cone Variability Factor 1 is introduced into p618 under control of the 49 nucleotide CMV enhancer/CMV-CBA hybrid promoter by insertion into the polylinker following digestion of p618 with the unique restriction en2ymes Notl and BamHI. Following this transgene insertion, the pro viral plasrnid was called p625.
  • Proviral plasmid p625 is co-transfected into a stable rep and cap expressing host packaging cell line B-50 (International Patent Application Publication No. WO 99/15685) with the adenovirus El, E2a, and E40RF6 DNA.
  • the gene expression cassette from the proviral plasmid is packaged into AAV particles employing iodixanol gradient purification followed by herparin-sepharose agarose column chromatography. Vector titers are determined using an infectious center assay.
  • AAV-hRdCVFl virus preparations are made and combined to a desired total volume.
  • the rAAV-hRdCVFl may be employed, for example, to transduce cultured target cells in vitro at multiplicities of infection (MOI) ranging from 10 3 to 10 6 rAAV viral particles per cell. Additionally, rAAV-hRdCVFl may be employed in vivo to transduce cells of the murine or other mammalian retina after administration by subretinal injection of 10"-10 13 viral particles. Expression of hRdCVFl in transduced cells or retinas is assessed by RT-PCR, immunocytochemistry, immunohistochemistry, and Western blot analysis, using conventional techniques.
  • MOI multiplicities of infection

Abstract

Proviral plasmids contain a modular gene expression cassette with one or a combination of (i) a wildtype 5' AAV2 ITR sequence flanked by unique restriction sites that permit ready removal or replacement of said ITR; (ii) a promoter flanked by unique restriction sites that permit ready removal or replacement of the entire promoter sequence; (iii) a polylinker sequence that permits insertion of a gene coding sequence without modification thereof, wherein the gene is operatively linked to, and under the regulatory control of, the aforementioned promoter; (iv) a bovine growth hormone polyadenylation sequence flanked by unique restriction sites that permit ready removal or replacement of said polyA sequence; and (v) a wildtype 3' AAV2 ITR sequence flanked by unique restriction sites that permit ready removal or replacement of the 3' ITR. These plasmids enable rapid manipulation of the components of the cassette, e.g., rapid mutation and/or replacement of any component, and thereby increase the efficiency of recombinant viral vector, e.g., rAAV, production.

Description

PRO VIRAL PLASMIDS FOR PRODUCTION OF RECOMBINANT
ADENO-AS SOCIATED VIRUS
INCORPORATION-BY-REFERENCE OF MATERIAL SUBMITTED IN
ELECTRONIC FORM
Applicant hereby incorporates by reference the Sequence Listing material filed in electronic form herewith. This file is labeled "UPN-X5873PCT_ST25.txt".
BACKGROUND OF THE INVENTION
Recombinant adeno-associated virus (rAAV) vectors have been developed for gene replacement therapy because they are non-pathogenic and exhibit a broad range of tissue specificity. These vectors generally retain the AAV inverted terminal repeats (ITRs) located at each end of a gene expression cassette, but lack the AAV rep and cap genes necessary for viral replication and packaging. Therefore rAAV cannot replicate, and viruses must be assembled in packaging cell lines with the rep and cap functions supplied in trans or expressed within the packaging cell itself. The gene expression cassette for insertion into the rAAV conventionally contains the therapeutic gene and the cis- regulatory elements including a promoter and a polyadenylation signal necessary for gene expression. In a conventional method of rAAV production, a gene expression cassette, located between the ITRs, is packaged in rAAV particles, which are then used in therapeutic applications.
The design and construction of the components, such as the plasmids and gene expression cassettes necessary for producing a recombinant AAV, can be quite labor intensive, due to the variety of plasmids and vectors available, and the need to modify the genes to fit into the appropriate plasmids. This complexity is further increased by pharmaceutical industry and governmental requirements that govern the process for obtaining approval of an rAAV for pharmaceutical use.
SUMMARY OF THE INVENTION
The invention described herein involves novel AAV proviral vectors characterized by ease of subcloning and which facilitate the ready substitution of alternative plasmid components. These vectors improve the efficiency of rAAV vector production and function. In one aspect, a proviral plasmid is provided in which all the functional elements of the proviral plasmids are modular and readily removable or replaceable by virtue of the unique flanking restriction sites. The modular aspect of these plasmids permits the expression of AAVs with different transgenes.
In another aspect, a proviral plasmid comprises a wildtype AAV2 ITR sequence, the ITR flanked by unique restriction sites that permit ready removal from the plasmid or replacement of the entire ITR. In one embodiment, the proviral plasmid comprises a 5' ITR and a 3' ITR as described above, each flanked by different unique restriction sites to permit removal or replacement of each ITR individually.
In another aspect, a proviral plasmid comprises a modular gene expression cassette comprising in operative association, (i) a wildtype 5' AAV2 ITR sequence flanked by unique restriction sites that permit ready removal or replacement of said ITR;
(ii) a CMV promoter comprising an upstream non-coding sequence of about 49 nucleotides of CMV to enhance the productivity of the promoter, flanked by unique restriction sites that permit ready removal or replacement of the entire promoter sequence;
(iii) a polylinker sequence that permits insertion of a gene coding sequence without modification thereof, wherein the gene is operatively linked to, and under the regulatory control of, the aforementioned promoter; (iv) a bovine growth hormone polyadenylation sequence flanked by unique restriction sites that permit ready removal or replacement of said polyA sequence; and (v) a wildtype 3' AAV2 ITR sequence flanked by unique restriction sites that permit ready removal or replacement of the 3' ITR. In one embodiment, this plasmid contains a cytomegalovirus (CMV) promoter and further comprises an upstream non-coding sequence of about 49 nucleotides of CMV to enhance the productivity of the promoter. In another embodiment, this plasmid contains a hybrid promoter comprising a CMV promoter sequence and a chicken beta actin (CBA) promoter sequence and further comprises an upstream non-coding sequence of about 49 nucleotides of CMV to enhance the productivity of the promoter. In this aspect, the entire promoter is flanked by unique restriction sites that permit ready removal and/or replacement of the enhancer/hybrid promoter sequence.
In another embodiment, the proviral plasmids described above further comprise a plasmid backbone comprising the elements necessary for replication in bacterial cells, and further comprise a kanamycin resistance gene (KanR), the plasmid backbone containing 5' and 3' transcriptional terminator/insulator sequences that isolate transcription in the backbone from transcription in the gene cassette. In another embodiment, the proviral plasmids described above further comprise a plasmid backbone comprising the elements necessary for replication in bacterial cells, and further comprise a kanamycin resistance gene ( anR), the plasmid backbone containing 5' and 3' transcriptional terminator/insulator sequences that isolate transcription in the backbone from transcription in the gene cassette and a lambda stuffer sequence.
In another aspect, a proviral plasmid is p604 of FIG.1 SEQ ID NO: 1.
In another aspect, a proviral plasmid is p617 of FIG. 2 SEQID NO: 2.
In another aspect, a proviral plasmid is p618 of FIG. 3 SEQ ID NO: 3.
In another aspect any of the proviral plasmids described above further comprises a gene encoding sequence inserted into the polylinker sequence.
In another aspect, a method is provided for generating a proviral plasmid as described herein.
In another aspect, a method is provided for generating a rAAV using any of the proviral plasmids described herein.
These and other embodiments and advantages of the invention are described in more detail below.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 is a map showing the features of the 4879 bp AAV proviral plasmid pAAV.CMV.eGFP.wtlTRs (p604). This plasmid contains a modular gene expression cassette in a plasmid backbone based on the pJ201 plasmid (DNA2.0, Menlo Park, CA). In the description below, the numbers in parentheses are the nucleotide number in SEQ ID NO: 1. The plasmid backbone contains a kanamycin resistance gene (9-803), pTR (1063- 1079), and nrn B1B2T1 txn terminator sequence (988-1162), a pTF3 (3535-3560), a bla txn terminator (3444-3744), an rpn txn terminator (3751-3864), the restriction site Aflll (4057) which may be employed for insertion of a stuffer and a pUC origin of replication (4036-4839). The gene expression cassette comprises in operative association:
(i) a synthetic 5' ITR (wildtype AAV 2) (1253-1382) which contains no deletions (1365-1382), the ITR flanked 5' by the unique restriction site Sail (1238) and 3' by the unique restriction site Nhel (1428);
(ii) a cytomegalovirus (CMV) promoter (1443-2018) comprising a 49 nucleotide CMV enhancer sequence extension, the CMV enhancer and minimal CMV promoter, the entire promoter flanked 5' by the unique restriction site Nhel (1428) and 3' by the unique restriction sites BstXI (2202) and Notl (2208), and the 49 nucleotide enhancer sequence flanked in a 239 nucleotide sequence by 5' Nhel (1428) and a 3' unique restriction site Ndel (1667);
(iii) a green fluorescent protein (eGFP) gene (2215-2937) flanked 5' by the unique restriction site BstXI (2202)or Notl (2208) and 3' by the unique restriction sites Hind III (2937), BamHI (2943) and Bglll (2952);
(iv) a bovine growth hormone polyadenylation signal (BGH poly A) (2952-3173), flanked 5* by unique restriction sites Hind III (2937), BamHI (2943) and Bglll (2952) and 3' by unique restriction site Xhol (3173) ; and
(v) a synthetic 3' ITR (wildtype AAV 2) (3221-3350) which contains no deletions, the ITR flanked 5' by unique restriction site Xhol (3173) and 3' by unique restriction site Bsu36I (3363).
FIG. 2 is a map showing the features of the 5060 bp AAV proviral plasmid pAAV.CMV.CBA.synlTR.short (p617). This plasmid contains a modular gene expression cassette in a plasmid backbone based on the pJ201 plasmid (DNA2.0, Menlo Park, CA). In the description below, the numbers in parentheses are the nucleotide number in SEQ ID NO: 2. The plasmid backbone contains a kanamycin resistance gene (9-803), pTR (1063- 1079), rrn Bl B2 Tl txn terminator sequence (988-1162), a pTF3 (3716-3741), a bla txn terminator (3625-3925), an rpn txn terminator (3932-4045), the restriction site Aflll (4057) which may be employed for insertion of a staffer, and a pUC origin of replication (4217-5020). The gene expression cassette comprises in operative association:
(i) a synthetic 5' ITR (wildtype AAV 2) (1253-1382) which contains no deletions, the ITR flanked 5' by the unique restriction site Sail (1238) and 3' by the unique restriction site Nhel (1428);
(ii) a promoter comprising a hybrid CMV/CBA promoter (1443- 3104), derived from the pDRIVE CAG plasmid (Invivogen, San Diego, CA) with an upstream extension of about 49 nucleotides of CMV enhancer; the entire promoter sequence including the upstream sequence is flanked 5' by the unique restriction site Nhel (1428) and 3' by the unique restriction sites Notl (3107), BamHI (3124), Pstl (3133) and Bglll (3133); the enhancer sequence extension is contained within a 239 nucleotide sequence as described for p604;
(iii) a polylinker sequence containing unique restriction sites Notl (3107), BamHI (3124), Pstl (3133), and Bglll (3133); (iv) a bovine growth hormone polyadenylation signal (BGH poly A) (3133-3354), flanked 5' by unique restriction sites Notl (3107), BamHI (3124), Pstl (3133) and Bglll (3133); and 3' by unique restriction site Xhol (3354);
(v) a synthetic 3' ITR (wildtype AAV 2) (3402-3531) which contains no deletions, the ITR flanked 5' by unique restriction site Xhol (3354) and 3' by unique restriction site Bsu36I (3544).
FIG. 3 is a map showing the features of the 10131 bp AAV pro viral plasmid pAAV.CMV.CBA.synlTR.long.stuffer 15 or p618 (SEQ ID NO: 3). In the description below, the numbers in parentheses are the nucleotide numbers in SEQ ID NO: 3. This plasmid contains a modular gene expression cassette in a plasmid backbone based on the pJ201 plasmid (DNA2.0, Menlo Park, CA). The plasmid backbone contains a kanamycin resistance gene (9-803), pTR (1063-1079), rrn Bl B2 Tl txn terminator sequence (988- 1 162), a pTF3 (3716-3741), a bla txn terminator (3625-3925), an rpn txn terminator (3932- 4045), a lambda stuffer sequence (4061-9127) which is inserted into the site Aflll, and a pUC origin of replication (9288-10091).
The gene expression cassette comprises in operative association:
(i) a synthetic 5' ITR (wildtype AAV 2) which contains no deletions, the ITR (1253 to 1382) flanked 5' by the unique restriction site Sail (1238) and 3' by the unique restriction site Nhel (1428);
(ii) a promoter comprising a hybrid CMV/CBA promoter, derived from the pDRIVE CAG plasmid (Invivogen, San Diego, CA) with an upstream extension of about 49 nucleotides of CMV enhancer, the entire promoter (1443-3104) flanked 5' by the unique restriction site Nhel (1428) and 3' by the unique restriction sites Notl (3107), Pstl (3133) and Bglll (3133); the enhancer sequence extension is contained within a 239 nucleotide sequence as described for p604;
(iii) a polylinker sequence (3105-3137) containing unique restriction sites , Notl (3107), Bglll (3133), and Pstl (3133);
(iv) a bovine growth hormone polyadenylation signal (BGH poly A) (3133-3354), flanked 5' by unique restriction sites BamHI (2943), Bglll (2952), Notl (3107), and Pstl (3133) and 3' by unique restriction site Xhol (3354); and
(v) a synthetic 3' ITR (wildtype AAV 2) which contains no deletions (3402-3531) flanked 5' by unique restriction site Xhol (3354) and 3' by unique restriction site Bsu36I (3544). FIG. 4 is a cartoon of the synthesized Exon 1 ORF of hRdCVFl (DNA2.0) and the modifications made to it for insertion into a proviral plasmid. The modifications include the addition of Notl and BamHI restriction sites, a stop codon embedded in Bell site to facilitate addition of epitope tag, and a complete Kozak consensus sequence that overlaps Notl. Only the hRdCVFl ORF of SEQ ID NO:4 is included as the transgene. The entire sequence depicts 357 bp.
FIG. 5 is a cartoon of a genomic fragment of hRdCVFl containing the Exon 1 ORF, exonic sequence upstream of the start codon, and intronic sequence downstream of stop codon. Modifications incorporated during synthesis (DNA2.0) include the addition of Notl and BamHI restriction sites. The entire sequence depicts 470bp.
FIG. 6 is a gel showing the results of a Western blot analysis of hRdCVFl expression in cultured cells after transfection, as described in Example 2.
FIG. 7 is a sequence showing the alignment of hRdCVFl genomic fragment of 456 bases (top line, SEQ ID NO: 4) and open reading frame or exon 1 ORF of 327 bases (bottom line; SEQ ID NO: 5).
DETAILED DESCRIPTION OF THE INVENTION
The novel AAV proviral plasmids described herein can be used to increase the productivity and efficiency of generating a variety of rAAV expressing a variety of targeted genes. All the functional elements of the proviral plasmids are modular and readily removable or replaceable by virtue of the unique flanking restriction sites. The modular aspect of these plasmids permits the efficient generation of AAVs which express different genes.
/. The Proviral Plasmids
As described herein, a proviral plasmid is designed to include a plasmid
"backbone" and a "minigene" or "gene expression cassette" flanked by AAV ITRs.
Together the ITRs and minigene comprise the recombinant AAV (rAAV) genome that is packaged into AAV particles.
As discussed in detail herein, the gene expression cassette includes, at a minimum, a transgene (i.e., the gene desired to be transported by the rAAV and expressed in selected cells) and its regulatory sequences, flanked by 5 ' and 3 ' AAV inverted terminal repeats (ITRs). A. Unique Restriction Sites
Every significant component in the rAAV genome contained within the proviral plasmid is flanked by unique restriction sites, i.e., restriction sites used only once in the plasmids to enable ready removal and/or replacement of individual components in the rAAV genome. By "unique restriction site" is meant a restriction site that is cleaved by an enzyme that cannot cleave another restriction site in the proviral plasmid. In one embodiment, the unique flanking restriction enzyme sites allow directional cloning of one or more components into the plasmid. Suitably, a pair of unique restriction enzyme sites flanking one component allows digestion at only a single locus in the proviral plasmid and ready insertion or deletion of only one component of the recombinant AAV genome.
In the present application, suitable restriction enzymes for unique use in the desired plasmids include those identified in the description of the figures and in the figures themselves. Suitable restriction enzymes may be identified using information readily available to those of skill in the art in the literature and in a variety of on-line databases, e.g., the REBASE™ database. Suitable restriction enzymes for the use in generating a proviral plasmid of this invention can be readily determined using a variety of computer programs and/or on-line databases. Suitable restriction enzymes are available from a variety of commercial sources including, e.g., New England Biolabs, Obiogene, Lift Technology, Roche, Clontech, Stratagene, Amersham, Pharmacia, among others.
B. Inverted Terminal Repeats of the Minigene
In one embodiment described herein, a proviral plasmid contains a wildtype ITR of AAV serotype 2. The ITRs are synthesized to remove a common mutation, i.e., a deletion in the upstream ITR that is present in many known vectors. This attribute alone allows use of these plasmids to improve packaging efficiency of rAAV during production. Without wishing to be bound by theory, the inventors theorized that mutated ITR sequences are corrected in the cell during production of the rAAV. However, use of the wildtype ITR, particularly the 5' ITR conserves the cell's resources and is more efficient. However, ITR sequences from other suitable AAV serotype sources may be selected or ITR mutations may be introduced by design to alter function of the rAAV.
In one embodiment, a significant attribute of these plasmids is that the intact wildtype ITR sequence is flanked by unique restriction sites that permit ready removal or replacement of the ITR from the plasmid. Thus, in one embodiment, the proviral plasmid comprises a 5' ITR flanked by unique restriction sites to permit its removal or replacement. In another embodiment, the proviral plasmid comprises a 3' ITR flanked by unique restriction sites to permit its removal or replacement. In another embodiment, the proviral plasmid contains a 5' ITR and a 3' ITR, each ITR flanked by unique restriction sites, so that each ITR may be individually removed from the plasmid, if desired. The proviral plasmids described in the FIGs. 1 -3 have identified exemplary ITRs.
This modular capacity of the ITRs in the proviral plasmids allows for ready insertion and replacement of a variety of ITR sequences and permits use of the proviral plasmids in a number of methods for generating recombinant viruses.
C. The Regulatory Sequences of the Minigene
These proviral plasmids also include regulatory elements which are operably linked to the transgene in a manner which permits the transgene's transcription, translation and/or expression in a cell transfected with the plasmid vector or infected with the virus produced generated by use of the proviral plasmids. As used herein, "operably linked" sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation (polyA) signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e. , Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance secretion of the encoded product. A great number of expression control sequences, including promoters which are native, constitutive, inducible and/or tissue-specific, are known in the art and may be utilized.
These proviral plasmids are designed so that the promoter and poly A sequences are modular and may be readily replaced with other known promoter and polyA sequences. In one embodiment, e.g., p604 described in detail in FIG. 1 , the promoter is a cytomegalovirus promoter (p604) comprising an enhancer and minimal promoter with a 49 nucleotide upstream extension of the enhancer sequence
5'-tagtaatcaattacggggtcattagttcatagcccatatatggagttcc-3' (SEQ ID NO: 6). Two unique restriction sites flank a 239 bp promoter fragment that contains this promoter/enhancer extension. Thus, this promoter enhancer/extension, is in fact, modular. One would remove the 239 bp fragment and replace with one without the extension to reduce the power of the promoter.
In another embodiment, the promoter is a hybrid promoter comprising a CMV enhancer sequence and a chicken beta actin (CBA) promoter sequence (the pDRIVE-CAG promoter (Invitrogen). In still another embodiment, the hybrid CMV/CBA promoter further comprises the upstream non-coding sequence of about 49 nucleotides of CMV SEQ ID NO: 6 to enhance the productivity of the promoter, wherein the upstream CMV enhancer/extension sequence is in the larger 239 sequence, discussed above is flanked by unique restriction sites that permit ready removal or replacement of the upstream CMV sequence from the hybrid promoter sequence. In the exemplary plasmids described in detail in FIGs 1-3, these promoter sequences are identified. It was surprisingly determined that the CMV/CBA hybrid promoter (Invivogen) was about twice as strong a promoter when it is preceded by about a sequence of about 49 nucleotides upstream of the CMV promoter sequence. In the proviral plasmids, this upstream enhancer sequence is flanked by unique restriction sites, permitting its removal and/or replacement should the strong promoter be too strong in expressing any particular transgene in the selected host cell. For example in the three exemplified plasmids, the 49 nucleotide extension SEQ ID NO: 6 is flanked 5' by unique restriction site Nhel and further downstream to create the 239 nucleotide sequence by unique restriction site Ndel.
Thus while the identified promoters are currently desirable, the proviral plasmids allow for the easy removal or replacement of the upstream CMV enhancer sequence alone by cleavage at the unique restriction sites flanking the enhancer sequence. In another aspect, the proviral plasmids permit removal or replacement of the entire
promoter/enhancer sequence by the presence of unique restriction sites at both ends of the entire promoter sequence.
Should replacement of a promoter sequence be desired these proviral plasmids allow for excision of the existing promoters and insertion between the unique sites of any number of conventionally employed promoters. Suitable constitutive or inducible promoter sequences are available from a variety of commercial sources, including, without limitation, Invitrogen, Clontech and Ariad. Many other systems, e.g., native promoters, tissue-specific promoters, etc., have been described and can be readily selected by one of skill in the art. See, e.g. , the promoters identified in US Patent No. 7,906, 111, incorporated by reference herein.
Another modular regulatory sequence in the proviral plasmids described herein is the polyadenylation sequence. In the exemplary plasmids of FIGs 1-3, the bovine growth hormone polyA site is flanked by unique restriction sites, thereby permitting its ready replacement or removal, as desired. The selection of the particular polyA sequence is not critical. It is a characteristic of these plasmids that quick replacement of this regulatory sequence is possible depending upon the selection of transgene, promoter and/or host cell.
Still other conventional regulatory sequences may be incorporated into the proviral plasmids, as desired.
D. The Poly-Linker Sequence and Transgene of the Minigene
The proviral plasmids described herein also contain a multiple cloning site/polylinker sequence for ready insertion of almost any transgene without the requirement for extensive modification of the transgene. In one embodiment this is due to the inclusion of a number of unique restriction sites, such as Notl, BamHI, Pstl and Bglll. In FIGs. 2 - 3 for plasmids p617 and p618, which plasmids have no transgenes, the polylinker sequences are located between the promoter sequence and the polyA sequence. Once a transgene is selected, the transgene (or a portion thereof, if it is a large transgene), may be inserted into the polylinker site of these plasmids.
The transgene is a nucleic acid sequence, heterologous to the ITR and regulatory sequences flanking the transgene, which encodes a polypeptide, protein, RNA, or other product of interest. The nucleic acid coding sequence is operatively linked to regulatory components in a manner which permits transgene transcription, translation, and/or expression in a host cell.
The composition of the transgene sequence will depend upon the use to which the resulting vector will be put. For example, one type of transgene sequence includes a reporter sequence, which upon expression produces a detectable signal. In one embodiment, a reporter sequence is enhanced GFP (eGFP). In p604, for example, the eGFP gene may be easily removed and replaced by any gene up to about 3.3 kb in length for expression in rAAV using current packaging protocols. In other embodiments, such reporter sequences include, without limitation, DNA sequences encoding β-lactamase, β- galactosidase (LacZ), alkaline phosphatase, thymidine kinase, green fluorescent protein (GFP), chloramphenicol acetyltransferase (CAT), luciferase, membrane bound proteins including, for example, CD2, CD4, CD8, the influenza hemagglutinin protein, and others well known in the art, to which high affinity antibodies directed thereto exist or can be produced by conventional means, and fusion proteins comprising a membrane bound protein appropriately fused to an antigen tag domain from, among others, hemagglutinin or Myc.
These coding sequences, when associated with regulatory elements which drive their expression, provide signals detectable by conventional means, including enzymatic, radiographic, colorimetric, fluorescence or other spectrographic assays, fluorescent activating cell sorting assays and immunological assays, including enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA) and immunohistochemistry. For example, where the marker sequence is the LacZ gene, the presence of the vector carrying the signal is detected by assays for beta-galactosidase activity. Where the transgene is green fluorescent protein or luciferase, the vector carrying the signal may be measured visually by color or light production in a luminometer.
However, desirably, the transgene is a non-marker sequence encoding a product which is useful in biology and medicine, such as proteins, peptides, RNA, enzymes, dominant negative mutants, or catalytic RNAs. Desirable RNA molecules include tRNA, dsRNA, ribosomal RNA, catalytic RNAs, siRNA, small hairpin RNA, trans-splicing RNA, and antisense RNAs. One example of a useful RNA sequence is a sequence which inhibits or extinguishes expression of a targeted nucleic acid sequence in the treated animal. Typically, suitable target sequences include oncologic targets and viral diseases. See, for examples of such targets the oncologic targets and viruses identified in US Patent No. 7,906,111, incorporated herein by reference. In one embodiment, the transgene is the human rod derived cone variability factor 1 (see e.g., NCBI Accession No. NT_01 1295.11 and NM_138454.1, incorporated by reference; SEQ ID NO: 4). In one embodiment, the transgene is exon 1 open reading frame (ORF) of the hRdCVFl cDNA, e.g., from the Ncol site at nucleotide 12 to the stop codon and flanked by Notl and BamHI restriction sites (SEQ ID NO: 5; FIG 4). In another embodiment, the transgene is the exon 1 ORF with exonic sequences upstream of the start codon and intronic sequences downstream of the stop codon flanked by the insertion of a Notl restriction site at nucleotide 3 of the hRdCVf exon 1 fragment and a BamHI restriction site at nucleotide position 466 of the fragment (FIG. 5). In one embodiment, the hRdCVf 1 exon 1 ORF is modified by embedding the stop codon in a Bell site at nucleotide position 341 to facilitate the addition of an epitope tag (FIG. 4). A complete Kozak consensus sequence was also included and overlaps the Notl sequence (FIG. 4).
The transgene may be used to correct or ameliorate gene deficiencies, which may include deficiencies in which normal genes are expressed at less than normal levels or deficiencies in which the functional gene product is not expressed. Alternatively, the transgene may provide a product to a cell which is not natively expressed in the cell type or in the host. A preferred type of transgene sequence encodes a therapeutic protein or polypeptide which is expressed in a host cell. The invention further includes using multiple transgenes. In certain situations, a different transgene may be used to encode each subunit of a protein, or to encode different peptides or proteins or portions of a single large protein. This is desirable when the size of the DNA encoding the protein subunit is large, e.g., for an immunoglobulin, the platelet-derived growth factor, or a dystrophin protein. Additionally this may be desirable for proteins encoded by genes that are mutated in inherited forms of blindness such as centrosomal protein 290kDa (CEP290) mutations in Leber Congenital Amaurosis and ATP-binding cassette, sub-family A member 4 (ABCR).mutations in Stargardt's Disease. In order for the cell to produce the multi- subunit protein, a cell is infected with the recombinant virus containing each of the different subunits. Alternatively, different subunits of a protein may be encoded by the same transgene. In one embodiment, a single transgene includes the DNA encoding each of the subunits, with the DNA for each subunit separated by an internal ribo2yme entry site (IRES). This is desirable when the size of the DNA encoding each of the subunits is small, e.g., the total size of the transgene expression cassette including DNA encoding the subunits and the IRES is less than 4.7 kilobases. As an alternative to an IRES, the DNA may be separated by sequences encoding a 2A peptide, which self-cleaves in a post- translational event. See, e.g., M.L. Donnelly, et al, J. Gen. Virol, 78(Pt 1): 13-21 (Jan 1997); Furler, S„ et al, Gene Ther., 8(11):864-873 (June 2001); Klump H., et al, Gene Ther., 8(10):811-817 (May 2001). This 2A peptide is significantly smaller than an IRES, making it well suited for use when space is a limiting factor. More often, when the transgene is large, consists of multi-subunits, or two transgenes are co-delivered, rAAV carrying the desired transgene(s) or subunits are co-administered to allow them to concatamerize in vivo to form a single vector genome. In such an embodiment, a first AAV may carry an expression cassette which expresses a single transgene and a second AAV may carry an expression cassette which expresses a different transgene for co- expression in the host cell. However, the selected transgene may encode any biologically active product or other product, e.g., a product desirable for study.
Suitable transgenes may be readily selected by one of skill in the art. The selection of the transgene is not considered to be a limitation of this invention, but may be selected from among many known transgenes (see, e.g., transgenes identified in US Patent No. 7,906,111, incorporated by reference herein). In embodiments, using the proviral plasmids in conventional packaging protocols, plasmid p604 can incorporate transgenes of up to 3.3 kb in length for expression in rAAV; and p617 and p618 can incorporate genes up to 2.4 kb in length. E. The Backbone of the Proviral Plasmids
In one embodiment, the plasmid backbone of the proviral plasmid includes the conventional elements necessary for replication and, optionally, integration in prokaryotic cells (e.g., bacterial cells). In one embodiment, the plasmid backbone is pJ201 (DNA2.0). See FIGs. 1-3. Still other backbones may be employed if desired expression is mammalian cells, or both.
In one embodiment, the backbone desirably contains a selectable marker, e.g., a kanamycin resistance gene (KanR). The selectable marker gene is located in the backbone of the proviral plasmid so that it will not be rescued and incorporated into the resulting recombinant virus when the proviral plasmid is used to produce rAAV. It can be used to signal the presence of the plasmids in bacterial cells. KanR is particularly useful in these proviral plasmids for the generation of rAAV which are clinically useful or for generation of rAAV used in the treatment of humans for diseases or disorders requiring rAAV- mediated delivery of a particular transgene.
In one embodiment, another optional component of the plasmid backbone is an origin of replication, such as the pUC origin of replication identified in FIGs. 1-3.
In another embodiment, the plasmid backbone contains 5' and 3' transcriptional terminator/insulator sequences that isolate transcription in the backbone from transcription in the gene cassette. Among such sequences exemplified in the FIGs. 1 -3 are pTR, nrn B1B2T1 txn terminator sequence, pTF3, a bla txn terminator, and an rpn txn terminator.
In another embodiment, the plasmid backbone further comprises a stuffer sequence, such as the stuffer sequence exemplified in p618 (see, FIGs. 2 and 3). A stuffer sequence is a non-coding sequence used to enlarge the backbone. The stuffer sequence contains no functional elements from its viral origin. It does not transcribe genes and contains no promoter. The stuffer, in the case of p618, is a 5.1 kb sequence derived from lambda phage. The stuffer increases the size of the proviral plasmid to greater than 8 kb. It prevents the backbone {i.e., all sequence outside of the recombinant AAV genome) from being packaged into the rAAV capsid, which has a capacity of only 4.7 kb.
Given this description of the proviral plasmids herein, one of skill in the art is expected to be able to select these and other plasmid backbone elements. Many such sequences are available (see, e.g., Sambrook et al, and references cited therein).
The modular proviral plasmids p604, p617 and p618, and variants thereof containing selected transgenes desirably incorporate a variety of the components described herein. p618 in particular is designed as a plasmid that can be used to make a vector for a clinical trial. These features include all of the necessary elements for expression and packaging, except the gene of interest. Any gene can be easily cloned into the multiple cloning site for cDNA that includes Notl, BamHI, Pstl, and BgHI. The use of these proviral plasmids as described herein improves the ease of subcloning transgenes of a variety of types and sizes into recombinant AAV genomes, facilitates the substitution of alternative plasmid features, and improves the efficiency of rAAV vector production and function. These plasmids may be made by resort to conventional techniques given the descriptions, including sequences provided herein. The method of making the proviral plasmids given this disclosure is within the skill of the art.
II. Generating Recombinant Viruses Using the Proviral Plasmids
The proviral plasmids may be employed in currently conventional packaging methodologies to generate a recombinant virus expressing the transgene carried by the proviral plasmids. Suitable production cell lines are readily selected by one of skill in the art. For example, a suitable host cell can be selected from any biological organism, including prokaryotic {e.g., bacterial) cells, and eukaryotic cells, including, insect cells, yeast cells and mammalian cells. Briefly, the proviral plasmid is transfected into a selected packaging cell, where it may exist transiently. Alternatively, the minigene or gene expression cassette with its flanking ITRs is stably integrated into the genome of the host cell, either chromosomally or as an episome. Suitable transfection techniques are known and may readily be utilized to deliver the recombinant AAV genome to the host cell. Typically, the proviral plasmids are cultured in the host cells which express the cap and/or rep proteins. In the host cells, the the gene expression cassettes with flanking AAV ITRs are rescued and packaged into the capsid protein or envelope protein to form an infectious viral particle.
Generally, when delivering the vector comprising the minigene by transfection, the proviral plasmid is delivered in an amount from about 5 μg to about 100 μg DNA, about 10 μg to about 50 g DNA to about 1 x 104 cells to about 1 x 1013 cells, or about 1 x 105 cells. However, the relative amounts of plasmid DNA to host cells may be adjusted, taking into consideration such factors as the selected proviral plasmid, the delivery method and the host cells selected.
To avoid undue repetition, reference is made throughout this description to the p618 plasmid. However, it should be appreciated that the other novel plasmids described herein can be constructed and used in a similar manner. The proviral plasmids described herein, are useful for a variety of purposes, but are particularly well suited for use in production of a recombinant adeno-associated virus containing the gene expression cassette. These plasmids and rAAV vectors, their elements, construction, and uses are known in the art and are described herein.
In one aspect, to generate a recombinant adeno-associated virus (AAV) containing a transgene carried by the p618 plasmid requires the AAV helper functions of the Rep and Cap proteins, and the adenoviral helper functions provided by the products of the adenovirus E2A, E4 and VA genes. In one method, the p618 plasmid is co-transfected into adenovirus-infected human embryonic kidney 293 (293) cells with a plasmid providing the AAV helper functions. In another method, 293 cells are transfected with three plasmids: p618 containing a transgene, a plasmid providing the AAV helper function, and a third plasmid that substitutes for the wild type (wt) adenovirus by providing E2A, E4 and VA adenoviral genes to enable viral replication. The second method offers the advantage of avoiding wt adenovirus infection and of yielding rAAV preparations that are presumed to be free of adenovirus. See, Shi et al, Virology J., 6:3 (January 2009).
The components required to be cultured in the host cell to package a recombinant AAV genome in an AAV capsid may be provided to the host cell in trans. Alternatively, any one or more of the required components (e.g., recombinant AAV genome, rep sequences, cap sequences, and/or helper functions) may be provided by a stable host cell which has been engineered to contain one or more of the required components using methods known to those of skill in the art. Most suitably, such a stable host cell will contain the required component(s) under the control of an inducible promoter. However, the required component(s) may be under the control of a constitutive promoter. Examples of suitable inducible and constitutive promoters are provided herein, in the discussion of regulatory elements suitable for use with the transgene.
In still another alternative, a selected stable host cell may contain selected component(s) under the control of a constitutive promoter and other selected component(s) under the control of one or more inducible promoters. For example, a stable host cell may be generated which is derived from 293 cells (which contain El helper functions under the control of a constitutive promoter), but which contains the rep and/or cap proteins under the control of inducible promoters. Still other stable host cells may be generated by one of skill in the art. The rep sequences, cap sequences, and helper functions required for producing the rAAV of the invention may be delivered to the packaging host cell in the form of any genetic element which transfer the sequences carried thereon. The selected genetic element may be delivered by any suitable method, including those described herein.
The methods used to construct any embodiment of this invention are known to those with skill in nucleic acid manipulation and include genetic engineering, recombinant engineering, and synthetic techniques. Such techniques include conventional cloning techniques of cDNA such as those described in texts (Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, NY.), use of overlapping oligonucleotide sequences of the virus genomes, polymerase chain reaction, and any suitable method which provides the desired nucleotide sequence. Standard transfection and co-transfection techniques are employed, e.g. , CaP04 precipitation techniques. Other conventional methods employed include homologous recombination of the viral genomes, plaquing of viruses in agar overlay, methods of measuring signal generation, and the like. See, e.g., K. Fisher et al, J. Virol, 70:520-532 (1993) and US Patent No. 5,478,745. Similarly, methods of generating rAAV virions are well known and the selection of a suitable method is not a limitation on the present invention.
The AAV and components described herein may be isolated or obtained from academic, commercial, or public sources (e.g., the American Type Culture Collection, Manassas, VA). Alternatively, the AAV sequences may be obtained through synthetic or other suitable means by reference to published sequences such as are available in the literature or in databases such as, e.g., GenBank®, PubMed®, or the like.
The host cell contains the sequences which drive expression of a novel AAV capsid protein of the invention (or a capsid protein comprising a fragment thereof) in the host cell and rep sequences of the same source as the source of the AAV ITRs found in the recombinant AAV genome, or a cross-complementing source. The AAV cap and rep sequences may be independently obtained from an AAV source as described above and may be introduced into the host cell in any manner known to one in the art as described above. Additionally, when pseudotyping an AAV vector, the sequences encoding each of the essential rep proteins may be supplied by different AAV sources {e.g., AAVl , AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8). For example, the rep78/68 sequences may be from AAV2, whereas the rep52/40 sequences may be from AAV8.
In one embodiment, the host cell stably contains the capsid protein under the control of a suitable promoter, such as those described above. Most desirably, in this embodiment, the capsid protein is expressed under the control of an inducible promoter. In another embodiment, the capsid protein is supplied to the host cell in trans. When delivered to the host cell in trans, the capsid protein may be delivered via a plasmid which contains the sequences necessary to direct expression of the selected capsid protein in the host cell. Most desirably, when delivered to the host cell in trans, the plasmid carrying the capsid protein also carries other sequences required for packaging the rAAV, e.g., the rep sequences.
In another embodiment, the host cell stably contains the rep sequences under the control of a suitable promoter, such as those described above. Most desirably, in this embodiment, the essential rep proteins are expressed under the control of an inducible promoter. In another embodiment, the rep proteins are supplied to the host cell in trans. When delivered to the host cell in trans, the rep proteins may be delivered via a plasmid which contains the sequences necessary to direct expression of the selected rep proteins in the host cell. Most desirably, when delivered to the host cell in trans, the plasmid carrying the capsid protein also carries other sequences required for packaging the rAAV, e.g., the rep and cap sequences.
Thus, in one embodiment, the rep and cap sequences may be transfected into the host cell on a single nucleic acid molecule and exist stably in the cell as an episome. In another embodiment, the rep and cap sequences are stably integrated into the chromosome of the cell. Another embodiment has the rep and cap sequences transiently expressed in the host cell. For example, a useful nucleic acid molecule for such transfection comprises, from 5' to 3', a promoter, an optional spacer interposed between the promoter and the start site of the rep gene sequence, an AAV rep gene sequence, and an AAV cap gene sequence.
Optionally, the rep and/or cap sequences may be supplied on a vector that contains other DNA sequences that are to be introduced into the host cells. For instance, the vector may contain the rAAV construct comprising the minigene. The vector may comprise one or more of the genes encoding the helper functions, e.g., the adenoviral proteins El , E2a, and E4 ORF6, and the gene for VAI RNA.
Preferably, the promoter used in this construct may be any of the constitutive, inducible or native promoters known to one of skill in the art or as discussed above. The selection of the AAV to provide any of these sequences does not limit the invention.
In another preferred embodiment, the promoter for rep is an inducible promoter, such as are discussed above in connection with the transgene regulatory elements. One preferred promoter for rep expression is the T7 promoter. The vector comprising the rep gene regulated by the T7 promoter and the cap gene, is transfected or transformed into a cell which either constitutively or inducibly expresses the T7 polymerase. See
International Patent Publication No. WO 98/10088, published March 12, 1998.
The spacer is an optional element in the design of the vector. The spacer is a DNA sequence interposed between the promoter and the rep gene ATG start site. The spacer may have any desired design; that is, it may be a random sequence of nucleotides, or alternatively, it may encode a gene product, such as a marker gene. The spacer may contain genes which typically incorporate start/stop and polyA sites. The spacer may be a non-coding DNA sequence from a prokaryote or eukaryote, a repetitive non-coding sequence, a coding sequence without transcriptional controls or a coding sequence with transcriptional controls. Two exemplary sources of spacer sequences are the phage ladder sequences or yeast ladder sequences, which are available commercially, e.g. , from Gibco or Invitrogen, among others. The spacer may be of any size sufficient to reduce expression of the rep78 and rep6E gene products, leaving the rep52, repAO and cap gene products expressed at normal levels. The length of the spacer may therefore range from about 10 bp to about 10.0 kbp, preferably in the range of about 100 bp to about 8.0 kbp. To reduce the possibility of recombination, the spacer is preferably less than 2 kbp in length; however, the invention is not so limited.
Although the molecule(s) providing rep and cap may exist in the host cell transiently (i.e., through transfection), it is preferred that one or both of the rep and cap proteins and the promoter(s) controlling their expression be stably expressed in the host cell, e.g., as an episome or by integration into the chromosome of the host cell. The methods employed for constructing embodiments of this invention are conventional genetic engineering or recombinant engineering techniques such as those described in the references above. While this specification provides illustrative examples of specific constructs, using the information provided herein, one of skill in the art may select and design other suitable constructs, using a choice of spacers, P5 promoters, and other elements, including at least one translational start and stop signal, and the optional addition of polyadenylation sites.
In another embodiment of this invention, the rep or cap protein may be provided stably by a host cell.
The packaging host cell also requires helper functions in order to package the rAAV of the invention. Optionally, these functions may be supplied by a herpesvirus. Most desirably, the necessary helper functions are each provided from a human or non- human primate adenovirus source, such as those described above and/or are available from a variety of sources, including the American Type Culture Collection (ATCC), Manassas, VA (US). In one currently preferred embodiment, the host cell is provided with and/or contains an Ela gene product, an Elb gene product, an E2a gene product, and/or an E4 ORF6 gene product. The host cell may contain other adenoviral genes such as VAI RNA, but these genes are not required. In a preferred embodiment, no other adenovirus genes or gene functions are present in the host cell.
By "adenoviral DNA which expresses the Ela gene product", it is meant any adenovirus sequence encoding Ela or any functional Ela portion. Adenoviral DNA which expresses the E2a gene product and adenoviral DNA which expresses the E4 ORF6 gene products are defined similarly. Also included are any alleles or other modifications of the adenoviral gene or functional portion thereof. Such modifications may be deliberately introduced by resort to conventional genetic engineering or mutagenic techniques to enhance the adenoviral function in some manner, as well as naturally occurring allelic variants thereof. Such modifications and methods for manipulating DNA to achieve these adenovirus gene functions are known to those of skill in the art.
The adenovirus Ela, Elb, E2a, and/or E40RF6 gene products, as well as any other desired helper functions, can be provided using any means that allows their expression in a cell. Each of the sequences encoding these products may be on a separate vector, or one or more genes may be on the same vector. The vector may be any vector known in the art or disclosed above, including plasmids, cosmids and viruses.
Introduction into the host cell of the vector may be achieved by any means known in the art or as disclosed above, including transfection, infection, electroporation, liposome delivery, membrane fusion techniques, high velocity DNA-coated pellets, viral infection and protoplast fusion, among others. One or more of the adenoviral genes may be stably integrated into the genome of the host cell, stably expressed as episomes, or expressed transiently. The gene products may all be expressed transiently, on an episome or stably integrated, or some of the gene products may be expressed stably while others are expressed transiently. Furthermore, the promoters for each of the adenoviral genes may be selected independently from a constitutive promoter, an inducible promoter or a native adenoviral promoter. The promoters may be regulated by a specific physiological state of the organism or cell (i.e., by the differentiation state or in replicating or quiescent cells) or by exogenously added factors, for example. The host cell itself may be selected from any biological organism, including prokaryotic (e.g., bacterial) cells, and eukaryotic cells, including, insect cells, yeast cells and mammalian cells. Particularly desirable host cells are selected from among any mammalian species, including, without limitation, cells such as A549, WEHI, 3T3, 10T1/2, BH , MDC , COS 1, COS 7, BSC 1, BSC 40, BMT 10, VERO, WI38, HeLa, 293 cells (which express functional adenoviral El), Saos, C2C12, L cells, HT1080, HepG2 and primary fibroblast, hepatocyte and myoblast cells derived from mammals including human, monkey, mouse, rat, rabbit, and hamster. The selection of the mammalian species providing the cells is not a limitation of this invention; nor is the type of mammalian cell, i.e., fibroblast, hepatocyte, tumor cell, etc. The requirements for the cell used is that it not carry any adenovirus gene other than El , E2a and/or E4 ORF6; it not contain any other virus gene which could result in homologous recombination of a contaminating virus during the production of rAAV; and it is capable of infection or transfection of DNA and expression of the transfected DNA. In a preferred embodiment, the host cell is one that has rep and cap stably transfected in the cell.
One host cell useful in the present invention is a host cell stably transformed with the sequences encoding rep and cap, and which is transfected with the adenovirus El , E2a, and E40RF6 DNA and a proviral plasmid carrying the recombinant AAV genome as described above. Stable rep and/or cap expressing cell lines, such as B-50 (International Patent Application Publication No. WO 99/15685), or those described in US Patent No. 5,658,785, may also be similarly employed. Another desirable host cell contains the minimum adenoviral DNA which is sufficient to express E4 ORF6. Yet other cell lines can be constructed using the novel AAV9 cap sequences of the invention.
The preparation of a host cell according to this invention involves techniques such as assembly of selected DNA sequences. This assembly may be accomplished utilizing conventional techniques. Such techniques include cDNA and genomic cloning, which are well known and are described in Sambrook et al., cited above, use of overlapping oligonucleotide sequences of the adenovirus and AAV genomes, combined with polymerase chain reaction, synthetic methods, and any other suitable methods which provide the desired nucleotide sequence.
Introduction of the proviral plasmids discussed herein and the other necessary elements into the host cell may also be accomplished using techniques known to the skilled artisan and as discussed throughout the specification. See, also, S. Shi et al, Virol. J., 6:3 (2009). In preferred embodiment, standard transfection techniques are used, e.g., CaP04 transfection or electroporation, and/or infection by hybrid adenoviras/AAV vectors into cell lines such as the human embryonic kidney cell line HEK 293 (a human kidney cell line containing functional adenovirus El genes which provides trans-acting El proteins).
The resulting recombinant AAV that express the product of the transgene is particularly well suited to gene delivery for therapeutic purposes and for immunization, including inducing protective immunity. Further, the compositions of the invention may also be used for production of a desired gene product in vitro. For in vitro production, a desired product {e.g., a protein) may be obtained from a desired culture following transduction of host cells with a rAAV containing the molecule encoding the desired product and culturing the cell culture under conditions which permit expression.
Alternatively a desired product may be obtained from a desired culture following transfection of host cells with proviral plasmid containing the desired transgene. The expressed product may then be purified and isolated, as desired. Suitable techniques for transfection, cell culturing, purification, and isolation are known to those of skill in the art.
The following examples are illustrative only, and do not limit the scope of the present invention.
EXAMPLE 1 : Construction of AA V proviral plasmids containing human RdCVFl
NCBI reference NT_011295.11, Homo sapiens chromosome 19 genomic contig, GRCh37.p2 reference primary assembly contains the nucleotedoxin-like protein 1 or rod- derived cone viability factor 1 (RdCVFl). The mRNA reference sequence is
NM_138454.1 (SEQ ID NO: 4).
A fragment including exon 1 with 5' UTR as well as upstream and downstream flanking intronic sequence, with a stop codon in the downstream intron is used. Exon 1 begins at nt 21 of the genomic fragment.
Based on the reference sequences, two DNA fragments comprising the open reading frame (ORF) and a genomic fragment are synthesized by DNA2.0. The latter is synthesized as a 470 bp fragment with 5' Notl and 3' BamHI sites to facilitate cloning. The 357 bp ORF cDNA is synthesized with a complete ozak consensus sequence that was partly embedded in a 5' Notl site. The stop codon is embedded in a Bell site, followed by a BamHI site. The Bell site is inserted to allow addition of epitope tags. Each fragment was synthesized by DNA2.0 and supplied in a plasmid vector pJ201. Each fragment (ORF and genomic fragment, SEQ ID NOs: 5 and 4, respectively) is subcloned into AAV proviral plasmids pAAV.CMV.CBA.synITR.short (p617), and pAAV.CMV.CBA.synITR.long (p618) and into an additional known plasmid. The resulting plasmids p623/ORF and p624/genomic fragment were derived from p617. The resulting plasmids p625/ORF and p626/genomic fragment are derived from plasmid p618.
EXAMPLE 2: Western Blot Assay
Plasmids included the newly made AAV-hRdCVFl proviral plasmids described in Example 1, as well as pAAV2.1 CMV.hRdCVFl (p368), another plasmid made by the inventors, and two pcDNA3-based plasmids p452 and p488 supplied by a third party.
To determine expression levels of RdCVFl from the plasmids, a Western blot experiment was performed as follows: 293T cells (passage number n+ 5) were seeded into 100 mm culture dishes. At 24 hours after seeding, the cells were approximately 30% confluent and were transfected with 25 μg/dish of endotoxin-free plasmid (Quiagen) using lipofectamine LTX (ratio 2.5 μΐ lipofectamine LTX per μg of plasmid).
The cell culture medium was DMEM with high glucose, no pyruvate, and 10% FBS (no antibiotic). At 6 h the medium was aspirated and replaced with 5 ml of serum- free Optimem medium per dish. 24 h post transfection, the conditioned media were collected and spun for 5 min at 1200 rpm to remove cells and debris. A protease inhibitor cocktail was added to the clarified media and they were stored at -80°C until use. Cells were scraped from each dish into PBS, and the collected cells were pooled with the cells collected from the conditioned medium. Each cell pellet was resuspended in 250 μΐ of RIPA containing protease inhibitor cocktail. After 1 h incubation on ice, DNA was cleared from each sample by passage of the lysate over glass beads. A Pierce Micro BCA kit was used to quantify protein, and 50 μg aliquots of cell lysate were made and frozen at -80°C.
On the day of the assay, conditioned media were thawed and concentrated at 4°C using Amicon Ultra- 15 concentrators with a 3 Kd cut-off. 5 ml of conditioned medium was concentrated to 400 μΐ volume (12.5x). Samples consisted of 50 μg cell lysate (in a volume of 21 μΐ) or 21 μΐ of concentrated, conditioned medium. Each sample represented roughly l/20th of the entire contents of the culture dish. Samples were denatured in lx LDS loading buffer (Invitrogen) with DTT for 10 min at 70°C and loaded onto NuPAGE 4-12% gradient Bis-Tris gels that were subsequently run in MES buffer (Invitrogen). Gels were electroblotted onto Hybond ECL nitrocellulose (Amersham) and the transfer was verified by Ponceau staining.
Blots were blocked ON at 4°C using 5% milk in PBS with 0.05% Tween-20, followed by ON incubation at 4°C in primary antibody (p51181 J67) diluted 1 :5000 in 3% milk in PBS-Tween-20. Incubation with the secondary antibody was 3 h at RT using a 1 : 15,000 dilution of donkey anti-rabbit Ig- HRP conjugated (Amersham ECL reagent). The HRP signal was detected using ECL-Plus reagent and a Typhoon scanner. Results are shown in FIG. 6.
As shown (FIG. 6A), all of the newly synthesized AAV proviral plasmids (lanes 2-7) as well as the inventors' additional plasmid (lane 8) express a strong band, between the 6 and 14 kd molecular weight markers that is detected by antibody p51 181 J67 and is consistent with the predicted size of 12.7 kd for hRdCVFl . The two pcDNA3-based plasmids provided by a third party (lanes 9 and 11) express a faint band that is slightly larger than the bands expressed by the other plasmids. Finally, plasmids p624 (lane 5) and p626 (lane 7) express a band just below the 17 kd molecular weight marker that most likely represents a product resulting from an incomplete translational stop at the predicted stop codon immediately after the ORF. Finally, plasmids p623, p624 (based on p617), p625 and p626 (based on p618) show increased expression of the 12.7 kd band relative to p621 and p622, indicating that the addition of upstream CMV sequences in the plasmids results in a more powerful promoter. In fact, the postulated read-through at the stop codon in plasmids containing the hRdCVFl genomic fragment is not seen in plasmid p622 (lane 3) which has a less powerful promoter than p624 (lane 5) and p626 (lane 7). Finally, there is no evidence of secretion of hRdCVFl in this experiment (FIG. 6B).
The best prospects for in vivo use are p625 and p626. The former is best for robust expression of a product of uniform length. The latter would be useful if the longer product is biologically relevant. These two p618-based plasmids are preferred over p623 and p624 that perform similarly, but do not contain a staffer in the plasmid backbone. These plasmids do not contain a deletion in the upstream ITR, and as judged by band intensities in the Western blots, the promoter appears to be twice as strong as the promoter in p621 and p622 which lacks the 49 nucleotide-long extension of the CMV enhancer.
EXAMPLE 3: Production of r AA V
As discussed in Example 2 above, human Rod-derived Cone Variability Factor 1 is introduced into p618 under control of the 49 nucleotide CMV enhancer/CMV-CBA hybrid promoter by insertion into the polylinker following digestion of p618 with the unique restriction en2ymes Notl and BamHI. Following this transgene insertion, the pro viral plasrnid was called p625.
Proviral plasmid p625 is co-transfected into a stable rep and cap expressing host packaging cell line B-50 (International Patent Application Publication No. WO 99/15685) with the adenovirus El, E2a, and E40RF6 DNA. The gene expression cassette from the proviral plasmid is packaged into AAV particles employing iodixanol gradient purification followed by herparin-sepharose agarose column chromatography. Vector titers are determined using an infectious center assay. AAV-hRdCVFl virus preparations are made and combined to a desired total volume.
The rAAV-hRdCVFl may be employed, for example, to transduce cultured target cells in vitro at multiplicities of infection (MOI) ranging from 103 to 106 rAAV viral particles per cell. Additionally, rAAV-hRdCVFl may be employed in vivo to transduce cells of the murine or other mammalian retina after administration by subretinal injection of 10"-1013 viral particles. Expression of hRdCVFl in transduced cells or retinas is assessed by RT-PCR, immunocytochemistry, immunohistochemistry, and Western blot analysis, using conventional techniques.
All publications and documents recited above, the Sequence Listing, and the entirety of US Provisional Patent Application No.: 61/486,608, filed May 16, 2011 , are incorporated herein by reference. Numerous modifications and variations are included in the scope of the above-identified specification and are expected to be obvious to one of skill in the art. Such modifications and alterations to the compositions and processes, such as selections of different AAV species and subtypes, are believed to be within the scope of the claims appended hereto.
TABLE 1
(Sequence Listing Free Text)
The following information is provided for sequences containing free text under numeric identifier <223>.
Figure imgf000026_0001

Claims

CLAIMS:
1. A proviral plasmid comprising a modular recombinant AAV genome comprising a wildtype AAV2 ITR sequence, the ITR flanked by unique restriction sites that permit ready removal or replacement of the ITR from the plasmid.
2. The plasmid according to claim 1 , further comprising a 5' ITR and a 3' ITR, each flanked by different unique restriction sites to permit removal or replacement of each ITR individually.
3. The plasmid according to claim 1 , wherein the modular recombinant AAV genome comprises, in operative association,
(i) a wildtype 5' AAV2 ITR sequence flanked by unique restriction sites that permit ready removal or replacement of said ITR;
(ii) a promoter flanked by unique restriction sites that permit ready removal or replacement of the entire promoter sequence;
(iii) a polylinker sequence that permits insertion of a gene coding sequence without modification thereof, wherein the gene is operatively linked to, and under the regulatory control of, the aforementioned promoter;
(iv) a bovine growth hormone polyadenylation sequence flanked by unique restriction sites that permit ready removal or replacement of said polyA sequence; and
(v) a wildtype 3' AAV2 ITR sequence flanked by unique restriction sites that permit ready removal or replacement of the 3' ITR.
4. The plasmid according to claim 3, wherein the promoter is a cytomegalovirus (CMV) promoter.
5. The plasmid according to claim 3, wherein the promoter is a hybrid promoter comprising a CMV promoter sequence and a chicken beta actin (CBA) promoter sequence.
6. The plasmid according to claim 5, wherein the hybrid CMV/CBA promoter further comprises an upstream non-coding sequence of about 49 nucleotides of CMV to enhance the productivity of the promoter, wherein the upstream CMV enhancer sequence is flanked by unique restriction sites that permit ready removal or replacement of the upstream CMV sequence from the hybrid promoter sequence.
7. The plasmids according to any of claims 1-6, further comprising a plasmid backbone comprising the elements necessary for replication in bacterial cells, and further comprising a kanamycin resistance gene ( anR).
8. The plasmid according to claim 7, further comprising 5' and 3' transcriptional terminator/insulator sequences that isolate transcription in the backbone from transcription in the gene cassette.
9. The plasmid according to claim 7, wherein the plasmid backbone further comprises a non-coding stuffer sequence that increases the backbone length and prevents reverse packaging of non-functional AAV genomes.
10. A proviral plasmid comprising:
(a) a modular recombinant AAV genome comprising in operative association:
(i) a wildtype 5' AAV2 ITR sequence flanked by unique restriction sites that permit ready removal or replacement of said ITR;
(ii) a hybrid promoter comprising a 49 nucleic acid cytomegalovirus sequence upstream of a cytomegalovirus (CMV)-chicken beta actin sequence, the hybrid promoter flanked by unique restriction sites that permit ready removal or replacement of the entire promoter sequence, and the upstream sequence flanked by unique restriction sites that permit ready removal or replacement of only the upstream CMV sequence, from the hybrid promoter sequence;
(iii) a multi-cloning polylinker sequence that permits insertion of a gene coding sequence without modification thereof, wherein the gene is operatively linked to, and under the regulatory control of, promoter (ii);
(iv) a bovine growth hormone polyadenylation sequence flanked by unique restriction sites that permit ready removal or replacement of said polyA sequence; and (v) a wildt pe 3' AAV2 ITR sequence flanked by unique restriction sites that permit ready removal or replacement of the 3' ITR; and
(b) a plasmid backbone comprising the elements necessary for replication in bacterial cells, and further comprising a kanamycin resistance gene, said plasmid backbone flanked by transcriptional terminator/insulator sequences that isolate transcription in the backbone from transcription in the gene cassette.
11. The plasmid according to claim 10, wherein said plasmid backbone (b) further comprises a non-coding lambda phage 5.1 kb stuffer sequence to increase backbone length and prevent reverse packaging of non-functional AAV genomes.
12. The plasmid according to any of claims 1-11, further comprising in the recombinant AAV genome a gene encoding sequence of up to 2.6 kb in length.
13. The plasmid according to any of claims 1-12, further comprising as the gene encoding sequence a nucleotide sequence encoding an open reading frame of human rod derived cone viability factor exon 1.
14. The plasmid according to any of claims 1-12, wherein the gene encoding sequence encodes a detectable reporter gene.
15. The plasmid according to claim 14, wherein the detectable reporter gene is selected from the group consisting of green fluorescent protein, red fluorescent protein, and beta-galactosidase.
16. The plasmid according to claim 1 comprising the structure of FIG. 1.
17. The plasmid according to claim 16 having the nucleotide sequence of SEQ ID NO: 1.
18. The plasmid according to claim 1 having the structure of FIG. 2.
19. The plasmid according to claim 18 having the nucleotide sequence of SEQ ID NO: 2.
20. The plasmid according to claim 1 having the structure of FIG. 3.
21. The plasmid according to claim 20 having the nucleotide sequence of SEQ ID NO: 3.
22. A cell culture comprising cells transfected with the plasmid according to any of claims 1 to 21.
23. A method of generating a rAAV virus comprising:
(a) obtaining a plasmid of any of claims 1-21 ;
(b) modifying the plasmid by removing or replacing at least one of an ITR sequence, the promoter or the upstream CMV sequence of the promoter, the BGH polyA sequence, or inserting a gene of interest into the polylinker, of said plasmid using the appropriate restriction enzymes;
(c) culturing a packaging cell carrying the modified in the presence of sufficient viral sequences to permit packaging of the gene expression cassette viral genome into an infectious AAV envelope or capsid.
24. A recombinant AAV produced according to the method of claim 23.
PCT/US2012/038063 2011-05-16 2012-05-16 Proviral plasmids for production of recombinant adeno-associated virus WO2012158757A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/117,312 US9249425B2 (en) 2011-05-16 2012-05-16 Proviral plasmids and production of recombinant adeno-associated virus
US14/979,786 US9896665B2 (en) 2011-05-16 2015-12-28 Proviral plasmids and production of recombinant adeno-associated virus

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161486608P 2011-05-16 2011-05-16
US61/486,608 2011-05-16

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/117,312 A-371-Of-International US9249425B2 (en) 2011-05-16 2012-05-16 Proviral plasmids and production of recombinant adeno-associated virus
US14/979,786 Continuation US9896665B2 (en) 2011-05-16 2015-12-28 Proviral plasmids and production of recombinant adeno-associated virus

Publications (1)

Publication Number Publication Date
WO2012158757A1 true WO2012158757A1 (en) 2012-11-22

Family

ID=46178803

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/038063 WO2012158757A1 (en) 2011-05-16 2012-05-16 Proviral plasmids for production of recombinant adeno-associated virus

Country Status (2)

Country Link
US (2) US9249425B2 (en)
WO (1) WO2012158757A1 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016183422A1 (en) 2015-05-14 2016-11-17 St. Jude Children's Research Hospital, Inc. Nucleic acid molecules containing spacers and methods of use thereof
WO2016185242A1 (en) 2015-05-21 2016-11-24 INSERM (Institut National de la Santé et de la Recherche Médicale) Synergistic combination of neuronal viability factors and uses thereof
WO2017106202A2 (en) 2015-12-14 2017-06-22 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
WO2018200542A1 (en) * 2017-04-24 2018-11-01 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
WO2018213618A1 (en) 2017-05-17 2018-11-22 The General Hospital Corporation Gene therapy for tuberous sclerosis
WO2018218359A1 (en) 2017-05-31 2018-12-06 The Trustees Of The University Of Pennsylvania Gene therapy for treating peroxisomal disorders
EP3361869A4 (en) * 2015-10-14 2019-04-03 Audentes Therapeutics, Inc. Nucleic acid molecules containing spacers and methods of use thereof
WO2019122403A1 (en) 2017-12-22 2019-06-27 Sparingvision Constructs comprising neuronal viability factors and uses thereof
WO2020079464A1 (en) 2018-10-17 2020-04-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for treating retinal degeneration disease by administering nucleolin polynucleotide or polypeptide
WO2021119053A1 (en) 2019-12-10 2021-06-17 Shire Human Genetic Therapies, Inc. Adeno associated virus vectors for the treatment of hunter disease
EP3746560A4 (en) * 2018-02-02 2021-12-01 University of Massachusetts Campaign-ready series of recombinant adeno-associated virus (raav) complementing plasmids
US11197936B2 (en) 2016-07-08 2021-12-14 The Trustees Of The University Of Pennsylvania Methods and compositions for treatment of disorders and diseases involving RDH12
WO2022076582A1 (en) 2020-10-07 2022-04-14 Regenxbio Inc. Gene therapy for ocular manifestations of cln2 disease
WO2022165313A1 (en) 2021-02-01 2022-08-04 Regenxbio Inc. Gene therapy for neuronal ceroid lipofuscinoses
US11427835B2 (en) 2013-03-15 2022-08-30 The Children's Hospital Of Philadelphia Vectors comprising stuffer/filler polynucleotide sequences and methods of use
WO2022183052A1 (en) 2021-02-26 2022-09-01 Takeda Pharmaceutical Company Limited Composition and methods for the treatment of fabry disease
WO2022223644A2 (en) 2021-04-20 2022-10-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Compositions and methods for treating retinal degenerative disorders
US11564996B2 (en) 2017-03-01 2023-01-31 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
US11591614B2 (en) 2017-05-11 2023-02-28 The Trustees Of The University Of Pennsylvania Gene therapy for ceroid lipofuscinoses
WO2023187728A1 (en) 2022-04-01 2023-10-05 Takeda Pharmaceutical Company Limited Gene therapy for diseases with cns manifestations
US11827898B2 (en) 2017-06-14 2023-11-28 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
EP4357359A1 (en) 2022-10-20 2024-04-24 Sparingvision G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd)
WO2024084075A1 (en) 2022-10-20 2024-04-25 Sparingvision Compositions and methods for treating retinal degenerative disorders

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012158757A1 (en) 2011-05-16 2012-11-22 The Trustees Of The University Of Pennsylvania Proviral plasmids for production of recombinant adeno-associated virus
US10577627B2 (en) 2014-06-09 2020-03-03 Voyager Therapeutics, Inc. Chimeric capsids
WO2015196179A1 (en) * 2014-06-20 2015-12-23 University Of Florida Research Foundation, Inc. Methods of packaging multiple adeno-associated virus vectors
WO2016005004A1 (en) * 2014-07-11 2016-01-14 Biontech Rna Pharmaceuticals Gmbh Stabilization of poly(a) sequence encoding dna sequences
CN112553229A (en) 2014-11-05 2021-03-26 沃雅戈治疗公司 AADC polynucleotides for treating Parkinson's disease
DK3218386T3 (en) 2014-11-14 2021-06-07 Voyager Therapeutics Inc MODULATORY POLYNUCLEOTID
KR102599909B1 (en) 2014-11-14 2023-11-09 보이저 테라퓨틱스, 인크. Compositions and methods of treating amyotrophic lateral sclerosis (als)
US11697825B2 (en) 2014-12-12 2023-07-11 Voyager Therapeutics, Inc. Compositions and methods for the production of scAAV
WO2017087900A1 (en) * 2015-11-19 2017-05-26 The Trustees Of The University Of Pennsylvania Compositions and methods for correction of heritable ocular disease
US10857240B2 (en) 2016-01-05 2020-12-08 The Trustees Of The University Of Pennsylvania Methods and compositions for treatment of ocular disorders and blinding diseases
US11299751B2 (en) 2016-04-29 2022-04-12 Voyager Therapeutics, Inc. Compositions for the treatment of disease
WO2017189959A1 (en) 2016-04-29 2017-11-02 Voyager Therapeutics, Inc. Compositions for the treatment of disease
CA3024449A1 (en) 2016-05-18 2017-11-23 Voyager Therapeutics, Inc. Compositions and methods of treating huntington's disease
AU2017267665C1 (en) 2016-05-18 2023-10-05 Voyager Therapeutics, Inc. Modulatory polynucleotides
AU2017321488B2 (en) 2016-08-30 2022-10-20 The Regents Of The University Of California Methods for biomedical targeting and delivery and devices and systems for practicing the same
US11603542B2 (en) 2017-05-05 2023-03-14 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
AU2018261003A1 (en) 2017-05-05 2019-11-14 Voyager Therapeutics, Inc. Compositions and methods of treating Huntington's Disease
JOP20190269A1 (en) 2017-06-15 2019-11-20 Voyager Therapeutics Inc Aadc polynucleotides for the treatment of parkinson's disease
AU2018302016A1 (en) 2017-07-17 2020-02-06 The Regents Of The University Of California Trajectory array guide system
TWI804518B (en) 2017-10-16 2023-06-11 美商航海家醫療公司 Treatment of amyotrophic lateral sclerosis (als)
WO2019079242A1 (en) 2017-10-16 2019-04-25 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
EP3790960A4 (en) * 2018-05-07 2022-02-23 Spark Therapeutics, Inc. Plasmid free aav vector producing cell lines
SG11202109554XA (en) * 2019-03-04 2021-09-29 Univ Pennsylvania Neuroprotective gene therapy targeting the akt pathway

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002078516A2 (en) * 2001-03-30 2002-10-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of cancer
WO2006130581A2 (en) * 2005-05-31 2006-12-07 Avigen, Inc. Methods for delivering genes

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5756283A (en) * 1995-06-05 1998-05-26 The Trustees Of The University Of Pennsylvania Method for improved production of recombinant adeno-associated viruses for gene therapy
US6281010B1 (en) * 1995-06-05 2001-08-28 The Trustees Of The University Of Pennsylvania Adenovirus gene therapy vehicle and cell line
US6258595B1 (en) * 1999-03-18 2001-07-10 The Trustees Of The University Of Pennsylvania Compositions and methods for helper-free production of recombinant adeno-associated viruses
EP1381276A4 (en) 2001-04-13 2005-02-02 Univ Pennsylvania Method of treating or retarding the development of blindness
US20030003582A1 (en) * 2001-05-08 2003-01-02 Tranzyme, Inc. Trans-viral vector mediated gene transfer to the retina
WO2004009768A2 (en) * 2002-07-18 2004-01-29 Invitrogen Corporation Viral vectors containing recombination sites
WO2009091912A2 (en) * 2008-01-15 2009-07-23 Abbott Laboratories Improved mammalian expression vectors and uses thereof
BRPI0800957A2 (en) * 2008-04-04 2009-11-17 Univ Rio De Janeiro biological vectors comprising the max gene, their production method, max gene expression method in cells and cytoprotective gene therapy method
US20120244127A1 (en) * 2009-10-01 2012-09-27 The Trustees Of The University Of Pennsylvania AAV Vectors Expressing SEC10 for Treating Kidney Damage
WO2012158757A1 (en) 2011-05-16 2012-11-22 The Trustees Of The University Of Pennsylvania Proviral plasmids for production of recombinant adeno-associated virus

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002078516A2 (en) * 2001-03-30 2002-10-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of cancer
WO2006130581A2 (en) * 2005-05-31 2006-12-07 Avigen, Inc. Methods for delivering genes

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
ALEJANDRO B. BALAZS ET AL: "Antibody-based protection against HIV infection by vectored immunoprophylaxis", NATURE, vol. 481, no. 7379, 30 November 2011 (2011-11-30), pages 81 - 84, XP055032827, ISSN: 0028-0836, DOI: 10.1038/nature10660 *
BOULAIRE J ET AL: "Transcriptional targeting to brain cells: Engineering cell type-specific promoter containing cassettes for enhanced transgene expression", ADVANCED DRUG DELIVERY REVIEWS, ELSEVIER BV, AMSTERDAM, NL, vol. 61, no. 7-8, 2 July 2009 (2009-07-02), pages 589 - 602, XP026139407, ISSN: 0169-409X, [retrieved on 20090423], DOI: 10.1016/J.ADDR.2009.02.007 *
BROWNE MARYBETH ET AL: "Gene transfer of pigment epithelium-derived factor suppresses tumor growth and angiogenesis in a hepatoblastoma xenograft model.", PEDIATRIC RESEARCH SEP 2006 LNKD- PUBMED:16857775, vol. 60, no. 3, September 2006 (2006-09-01), pages 282 - 287, XP055035789, ISSN: 0031-3998 *
CHIKHLIKAR P ET AL: "Inverted terminal repeat sequences of adeno-associated virus enhance the antibody and CD8<+> responses to a HIV-1 p55Gag/LAMP DNA vaccine chimera", VIROLOGY, ACADEMIC PRESS,ORLANDO, US, vol. 323, no. 2, 1 June 2004 (2004-06-01), pages 220 - 232, XP004515297, ISSN: 0042-6822, DOI: 10.1016/J.VIROL.2004.02.025 *
CHO M A ET AL: "HoxD10 gene delivery using adenovirus/adeno-associate hybrid virus inhibits the proliferation and tumorigenicity of GH4 pituitary lactotrope tumor cells", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ACADEMIC PRESS INC. ORLANDO, FL, US, vol. 371, no. 3, 4 July 2008 (2008-07-04), pages 371 - 374, XP022670705, ISSN: 0006-291X, [retrieved on 20080428], DOI: 10.1016/J.BBRC.2008.04.085 *
KESSLER P D ET AL: "GENE DELICERY TO SKELETAL MUSCLE RESULTS IN SUSTAINED EXPRESSION AND SYSTEMIC DELIVERY OF A THERAPEUTIC PROTEIN", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, US, vol. 93, 1 November 1996 (1996-11-01), pages 14082 - 14087, XP002032009, ISSN: 0027-8424, DOI: 10.1073/PNAS.93.24.14082 *
LINGFEI XU ET AL: "CMV-[beta]-Actin Promoter Directs Higher Expression from an Adeno-Associated Viral Vector in the Liver than the Cytomegalovirus or Elongation Factor 1[alpha] Promoter and Results in Therapeutic Levels of Human Factor X in Mice", HUMAN GENE THERAPY, vol. 12, no. 5, 20 March 2001 (2001-03-20), pages 563 - 573, XP055035696, ISSN: 1043-0342, DOI: 10.1089/104303401300042500 *
LIU ET AL: "Specific and efficient transduction of cochlear inner hair cells with recombinant adeno-associated virus type 3 vector", MOLECULAR THERAPY, ACADEMIC PRESS, SAN DIEGO, CA, US, vol. 12, no. 4, 1 October 2005 (2005-10-01), pages 725 - 733, XP005078452, ISSN: 1525-0016, DOI: 10.1016/J.YMTHE.2005.03.021 *
SONG ET AL: "Islet cell differentiation in liver by combinatorial expression of transcription factors Neurogenin-3, BETA2, and RIPE3b1", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ACADEMIC PRESS INC. ORLANDO, FL, US, vol. 354, no. 2, 30 January 2007 (2007-01-30), pages 334 - 339, XP005737622, ISSN: 0006-291X, DOI: 10.1016/J.BBRC.2006.12.216 *
WANG C Y ET AL: "Astrocytic expression of transgene in the rat brain mediated by baculovirus vectors containing an astrocyte-specific promoter", GENE THERAPY, vol. 13, no. 20, October 2006 (2006-10-01), pages 1447 - 1456, ISSN: 0969-7128 *
WANG CHAO-YANG ET AL: "Adeno-associated virus inverted terminal repeats improve neuronal transgene expression mediated by baculoviral vectors in rat brain", HUMAN GENE THERAPY, vol. 16, no. 10, October 2005 (2005-10-01), pages 1219 - 1226, ISSN: 1043-0342 *
YAN ZIYING ET AL: "Inverted terminal repeat sequences are important for intermolecular recombination and circularization of adeno-associated virus genomes", JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 79, no. 1, 1 January 2005 (2005-01-01), pages 364 - 379, XP002399264, ISSN: 0022-538X *

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11427835B2 (en) 2013-03-15 2022-08-30 The Children's Hospital Of Philadelphia Vectors comprising stuffer/filler polynucleotide sequences and methods of use
WO2016183422A1 (en) 2015-05-14 2016-11-17 St. Jude Children's Research Hospital, Inc. Nucleic acid molecules containing spacers and methods of use thereof
JP2021184734A (en) * 2015-05-14 2021-12-09 セント ジュード チルドレンズ リサーチ ホスピタル インコーポレイテッド Nucleic acid molecules containing spacers and methods of use thereof
JP2018515102A (en) * 2015-05-14 2018-06-14 セント ジュード チルドレンズ リサーチ ホスピタル インコーポレイテッド Nucleic acid molecules containing spacers and methods of use thereof
AU2016260401B2 (en) * 2015-05-14 2022-05-19 St. Jude Children's Research Hospital, Inc. Nucleic acid molecules containing spacers and methods of use thereof
US11103597B2 (en) 2015-05-14 2021-08-31 St. Jude Children's Research Hospital, Inc. Nucleic acid molecules containing spacers outside ITR
JP7316326B2 (en) 2015-05-14 2023-07-27 セント ジュード チルドレンズ リサーチ ホスピタル インコーポレイテッド Nucleic acid molecules containing spacers and methods of their use
EP3294309A4 (en) * 2015-05-14 2019-01-16 St. Jude Children's Research Hospital, Inc. Nucleic acid molecules containing spacers and methods of use thereof
WO2016185242A1 (en) 2015-05-21 2016-11-24 INSERM (Institut National de la Santé et de la Recherche Médicale) Synergistic combination of neuronal viability factors and uses thereof
EP3361869A4 (en) * 2015-10-14 2019-04-03 Audentes Therapeutics, Inc. Nucleic acid molecules containing spacers and methods of use thereof
US11230720B2 (en) 2015-10-14 2022-01-25 Audentes Therapeutics, Inc. Nucleic acid molecules containing spacers and methods of use thereof
AU2016338565B2 (en) * 2015-10-14 2021-11-18 Audentes Therapeutics, Inc. Nucleic acid molecules containing spacers and methods of use thereof
EP3795180A1 (en) 2015-12-14 2021-03-24 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
US11090392B2 (en) 2015-12-14 2021-08-17 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
WO2017106202A2 (en) 2015-12-14 2017-06-22 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
US11197936B2 (en) 2016-07-08 2021-12-14 The Trustees Of The University Of Pennsylvania Methods and compositions for treatment of disorders and diseases involving RDH12
US11564996B2 (en) 2017-03-01 2023-01-31 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
US11879133B2 (en) 2017-04-24 2024-01-23 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
WO2018200542A1 (en) * 2017-04-24 2018-11-01 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
US11591614B2 (en) 2017-05-11 2023-02-28 The Trustees Of The University Of Pennsylvania Gene therapy for ceroid lipofuscinoses
WO2018213618A1 (en) 2017-05-17 2018-11-22 The General Hospital Corporation Gene therapy for tuberous sclerosis
WO2018218359A1 (en) 2017-05-31 2018-12-06 The Trustees Of The University Of Pennsylvania Gene therapy for treating peroxisomal disorders
US11793887B2 (en) 2017-05-31 2023-10-24 The Trustees Of The University Of Pennsylvania Gene therapy for treating peroxisomal disorders
US11827898B2 (en) 2017-06-14 2023-11-28 The Trustees Of The University Of Pennsylvania Gene therapy for ocular disorders
WO2019122403A1 (en) 2017-12-22 2019-06-27 Sparingvision Constructs comprising neuronal viability factors and uses thereof
EP3746560A4 (en) * 2018-02-02 2021-12-01 University of Massachusetts Campaign-ready series of recombinant adeno-associated virus (raav) complementing plasmids
US11884926B2 (en) 2018-02-02 2024-01-30 University Of Massachusetts Campaign-ready series of recombinant adeno-associated virus (rAAV) complementing plasmids
WO2020079464A1 (en) 2018-10-17 2020-04-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for treating retinal degeneration disease by administering nucleolin polynucleotide or polypeptide
WO2021119053A1 (en) 2019-12-10 2021-06-17 Shire Human Genetic Therapies, Inc. Adeno associated virus vectors for the treatment of hunter disease
WO2022076582A1 (en) 2020-10-07 2022-04-14 Regenxbio Inc. Gene therapy for ocular manifestations of cln2 disease
WO2022165313A1 (en) 2021-02-01 2022-08-04 Regenxbio Inc. Gene therapy for neuronal ceroid lipofuscinoses
WO2022183052A1 (en) 2021-02-26 2022-09-01 Takeda Pharmaceutical Company Limited Composition and methods for the treatment of fabry disease
WO2022223644A2 (en) 2021-04-20 2022-10-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Compositions and methods for treating retinal degenerative disorders
WO2023187728A1 (en) 2022-04-01 2023-10-05 Takeda Pharmaceutical Company Limited Gene therapy for diseases with cns manifestations
EP4357359A1 (en) 2022-10-20 2024-04-24 Sparingvision G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd)
WO2024084075A1 (en) 2022-10-20 2024-04-25 Sparingvision Compositions and methods for treating retinal degenerative disorders

Also Published As

Publication number Publication date
US20140087444A1 (en) 2014-03-27
US20160108373A1 (en) 2016-04-21
US9249425B2 (en) 2016-02-02
US9896665B2 (en) 2018-02-20

Similar Documents

Publication Publication Date Title
US9896665B2 (en) Proviral plasmids and production of recombinant adeno-associated virus
JP7444521B2 (en) A scalable method for producing recombinant adeno-associated virus (AAV) vectors in a serum-free suspension cell culture system suitable for clinical use.
JP5922095B2 (en) Pharmacologically induced transgene ablation system
US6936466B2 (en) Transcriptionally-activated AAV inverted terminal repeats (ITRs) for use with recombinant AAV vectors
US6953690B1 (en) Compositions and methods for helper-free production of recombinant adeno-associated viruses
KR20200033840A (en) Enhancers for improved cell transfection and / or rAAV vector production
AU2409200A (en) Compositions and methods useful for production of recombinant viruses which require helper viruses
EP1064393A1 (en) Compositions and methods for helper-free production of recombinant adeno-associated viruses
WO2003024502A2 (en) Compositions and methods useful for non-invasive delivery of therapeutic molecules to the bloodstream
WO2020086881A1 (en) Aav triple-plasmid system
Park et al. Scalable production of adeno‐associated virus type 2 vectors via suspension transfection
WO2021156609A1 (en) Process for making adenoassociated viral vectors
WO1999020773A2 (en) TRANSCRIPTIONALLY-ACTIVATED AAV INVERTED TERMINAL REPEATS (ITRs) FOR USE WITH RECOMBINANT AAV VECTORS
JP2003511037A (en) Production of recombinant AAV using adenovirus containing AAV rep / cap gene
US20220145328A1 (en) STABLE CELL LINES FOR INDUCIBLE PRODUCTION OF rAAV VIRIONS
WO2023172963A2 (en) Recombinant aav vectors and uses thereof
US20220242917A1 (en) Compositions and methods for producing adeno-associated viral vectors
KR20230145357A (en) Histidine-rich peptides as transformation reagents for rAAV and rBV production.
JP2023546116A (en) Nucleic acid constructs for VA RNA transcription
KR20240026174A (en) Methods for controlling adeno-associated virus production
JP2024501223A (en) Producer cells with low levels of VA-RNA
WO2022223954A1 (en) Dna amplification method using care elements

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12724461

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14117312

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12724461

Country of ref document: EP

Kind code of ref document: A1