AU722624B2 - An inducible method for production of recombinant adeno-associated viruses utilizing T7 polymerase - Google Patents

An inducible method for production of recombinant adeno-associated viruses utilizing T7 polymerase Download PDF

Info

Publication number
AU722624B2
AU722624B2 AU41833/97A AU4183397A AU722624B2 AU 722624 B2 AU722624 B2 AU 722624B2 AU 41833/97 A AU41833/97 A AU 41833/97A AU 4183397 A AU4183397 A AU 4183397A AU 722624 B2 AU722624 B2 AU 722624B2
Authority
AU
Australia
Prior art keywords
aav
host cell
vector
recombinant
rep
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU41833/97A
Other versions
AU4183397A (en
Inventor
Nancie Chen
James M. Wilson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Pennsylvania Penn
Original Assignee
University of Pennsylvania Penn
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Pennsylvania Penn filed Critical University of Pennsylvania Penn
Publication of AU4183397A publication Critical patent/AU4183397A/en
Application granted granted Critical
Publication of AU722624B2 publication Critical patent/AU722624B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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

Description

it 1I WO 98/10088 PCT/US97/15716 AN INDUCIBLE METHOD FOR PRODUCTION OF RECOMBINANT ADENO-ASSOCIATED VIRUSES UTILIZING T7 POLYMERASE Background of the Invention Adeno-associated virus is a replicationdeficient parvovirus, the genome of which is about 4.6 kb in length, including 145 nucleotide inverted terminal repeats (ITRs). The single-stranded DNA genome of AAV contains genes responsible for replication (rep) and formation of virions (cap).
When this nonpathogenic human virus infects a human cell, the viral genome integrates into chromosome 19 resulting in latent infection of the cell. Production of infectious virus and replication of the virus does not occur unless the cell is coinfected with a lytic helper virus such as adenovirus or herpesvirus. Upon infection with a helper virus, the AAV provirus is rescued and amplified, and both AAV and helper virus are produced.
AAV possesses unique features that make it attractive as a vector for delivering foreign DNA to cells. Various groups have studied the potential use of AAV in the treatment of disease states.
However, an obstacle to the use of AAV for delivery of DNA is the lack of highly efficient methods for encapsidation of recombinant genomes. See, R. Kotin, Hum. Gene Ther., 5:793-801 (1994). Furthermore, the rep gene product is toxic to cells and thus cannot be expressed at high levels. For example, previously known methods employ transfection of host cells with a rAAV genome which lacks rep and cap genes followed by coinfection with wild-type AAV and adenovirus. However, this method leads to unacceptably high levels of wildtype AAV. Incubation of cells with rAAV in the absence of contaminating wild-type AAV or helper adenovirus is associated with little recombinant gene expression. And, in the absence of the AAV rep gene product, integration is inefficient and not directed to chromosome 19.
Ih ft WO 98/10088 PCT/US97/15716 2 Bacteriophage T7 RNA polymerase (T7 Pol) is the product of T7 gene 1, which can recognize its responsive promoter sequence specifically and exhibit a high transcriptase activity Chamberlin et al, Nature, 228:227-231 (1970); J. Dunn and F. Studier, J. Mol.
Biol., 166:447-535 (1983); and B. Moffatt et al, Cell, 49:221-227 (1987)]. It has been used for heterologous expression of proteins in E. coli Tabor and C.
Richardson, Proc. Natl. Acad. Sci. USA, 82:1074-1078 (1985); F. Studier and B. Moffatt, J. Mol. Biol., 189:113-130 (1986)], in recombinant vaccinia virusinfected eukaryotic cells Fuerst et al, Proc. Natl.
Acad. Sci. USA, 83:8122-8126 (1986); A. Ramsey-Ewing and B. Moss, J. Biol. Chem., 271:16962-16966 (1996)], and in mammalian cells Lieber et al, Nucl. Acids Res., 17:8485-8493 (1989)].
What is needed is an efficient method for production of rAAV which avoids the problems associated with rep toxicity for the packaging cell.
Summary of the Invention The present invention provides an inducible method for efficient production of rAAV which makes use of T7 polymerase. T7 Pol is derived from lambda phage and its promoter is not active in mammalian cells. Thus, expression of rep/cap can be controlled by placing these genes under control of the T7 promoter and providing the T7 Pol in trans or under the control of an inducible promoter. Thus, this method avoids the toxic effects of rep which rendered prior art methods of producing rAAV inefficient. The method of the invention is particularly suitable for large scale production of rAAV, which is desired for rAAV vectors to be used in gene therapy.
In one aspect, the invention provides a method of producing rAAV which utilizes three vectors. A first WO 98/10088 PCTIUS97/15716 3 vector is capable of expressing T7 polymerase in the host cell following transfection or infection, A second vector comprises the AAV rep and cap genes under the control of T7 promoter sequences (T7/rep/cap). The third vector comprises a cassette consisting essentially of 5' and 3' AVV inverted terminal repeats (ITRs) flanking a selected transgene. A host cell containing these three vectors is cultured under conditions which permit replication and packaging of a recombinant AAV, and the rAAV is recovered.
In another aspect, the invention prpvides a method in which a host cell is stably transfected with one of the three components of the system used in the triple infection system. The remaining components are 15 introduced into the host cell, as described above.
SIn one embodiment, the invention provides a method in which a vector comprising T7 rep cap and a vector comprising a cassette consisting essentially of a selected minigene flanked by 5' and 3' AAV ITRs are introduced into a host cell expressing T7 polymerase. The host cell is then cultured under conditions which permit production of rAAV. In another embodiment, this invention provides a method which utilizes a host cell stably transfected with a plasmid containing T7/rep/cap. A vector 25 comprising T7 pol and a vector consisting essentially of a cassette comprising 5' AAV inverse terminal repeat (ITR), a selected minigene, and 3' AAV ITR are introduced into the host cell. The host cell is cultured under conditions which permit production of rAAV. In still another embodiment, the invention provides a method which utilizes a host cell stably transfected with a rescuable rAAV cassette. A vector comprising T7 pol and a vector comprising T7 rep cap are R4{- ontroduced into the host cell.
Ehost cell is cultured under conditions which permit 1V 35 piduction of rAAV.
/T
WO 98/10088 PCT/US97/15716 4 In yet another aspect, the present invention provides a method which utilizes a host cell stably transfected with two of the three components of the system used in the triple infection system. The remaining component is then introduced into the host cell, as described above.
In a further aspect, the present invention provides a method which utilizes a host cell stably transfected with the three components of the system used in the triple infection system. In this aspect, the T7 Pol is controlled by an inducible promoter.
In still a further aspect, the invention provides a rAAV produced according to the method of the invention.
Other aspects and advantages of the present invention are described further in the following detailed description of the preferred embodiments thereof.
Brief Description of the Drawings Fig. 1 provides a schematic illustration of the construction of a recombinant adenovirus containing the T7 polymerase gene.
Fig. 2 provides a schematic illustration of the construction of a recombinant plasmid containing the AAV rep/cap genes under control of a T7 promoter.
Fig. 3 provides a schematic illustration of the construction of a recombinant adenovirus containing the rep/cap genes under control of a T7 promoter.
Fig. 4 provides a schematic illustration of the construction of a recombinant hybrid Ad/AAV virus.
Detailed Description of the Invention The invention provides an inducible method for efficient production of recombinant AAV vectors useful particularly for gene delivery and transfer.
WO 98/10088 PCT/US97/15716 Specifically, the invention provides methods of AAV production in which expression of the toxic but necessary rep gene is controlled by the T7 promoter.
Thus, in one aspect, the method of the invention for production of rAAV involves introducing into a host cell the AAV rep and cap genes under control of a T7 promoter, and a recombinant adeno-associated virus (rAAV) cassette containing a selected minigene flanked by AAV ITRs. Upon introduction of a gene encoding T7 pol, high level expression of rep protein from the T7/rep/cap construct is induced and cells may be grown on a large scale. When rep expression is desired, the cells are caused to express the T7 polymerase which acts on the T7 promoter. This facilitates the efficient replication and packaging of rAAV carrying a gene of interest.
A host cell may be triple transfected (or infected) with vectors containing the above elements.
Alternatively, a host cell which expresses one or more of the required elements and may be transfected/infected with the remaining elements is utilized. In another alternative, a host cell is utilized which stably expresses all three elements of the system, and the T7 pol is placed under the control of an inducible promoter, which permits rep/cap expression to be controlled and the avoidance of toxic effects to the cell.
For each of the vector components used in the method of the invention, adenoviral constructs are currently preferred. However, using the information provided herein and known techniques, one of skill in the art could readily construct a different viral (adenoviral or non-adenoviral) or a plasmid vector which is capable of driving expression of the desired genes in the host cell. For example, although less preferred because of their inability to infect non-dividing cells, vectors WO 98/10088 PCT/US97/15716 6 carrying the required elements of this system, the T7 polymerase, may be readily constructed using retroviruses. Therefore, this invention is not limited by the virus or plasmid selected for purposes of introducing the T7 pol, T7/rep/cap, or AAV cassette into the host cell. Desirably, at least one of the vectors is a virus which provides the necessary helper functions to enable packaging. Alternatively, the helper functions may be provided by a co-transfected adenovirus or herpesvirus. Suitable techniques for introducing these vectors into the host cell are discussed below and are known to those of skill in the art. As used herein, a "host cell" is any cell (cell line), preferably mammalian, which permits expression of the T7 pol and T7/rep/cap and packaging of the rAAV containing the cassette, under the conditions described herein.
Suitable packaging cells are known, and may be readily selected by the skilled artisan.
A. Triple Infection/Transfection As stated above, a host cell used for assembly and packaging of recombinant AAV may be transfected with plasmid vectors or infected with viral vectors containing the required components of the system.
1. T7 Pol Vectors In a preferred embodiment, a first vector contains the T7 Pol gene under the control of a suitable promoter. In example 5 below, the nuclear localized T7 Pol gene is obtained from a publicly available plasmid Strauss, Nucleic Acid Res., 17:8485-8493 (1989)]. However, the gene may alternatively be obtained from other commercial and academic sources, including the American Type Culture Collection (pTF7-3, Accession No. 484944). See, also GenBank accession number M30308. Desirably, the T7 pol WO 98/10088 PCT/US97/15716 7 gene is linked to a nuclear localization signal, such as that described in Dunn, Gene, 68:259-266 (1988), using conventional techniques.
Desirably, T7 Pol is under the control of a cytomegalovirus (CMV) immediate early promoter/enhancer [see, Boshart et al, Cell, 41:521-530 (1985)]. However, other suitable promoters may be readily selected by one of skill in the art.
Useful promoters may be constitutive promoters or regulated (inducible) promoters, which will enable control of the amount of the transgene to be expressed.
For example, another suitable promoter includes, without limitation, the Rous sarcoma virus LTR promoter/enhancer.
Still other promoter/enhancer sequences may be selected by one of skill in the art.
In addition, the vector also includes other conventional regulatory elements necessary to drive expression of T7 Pol in a cell transfected with the vector. Such regulatory elements are known to those of skill in the art.
2. T7/Rep/Cap Vectors The second vector component of this system contains the rep and cap genes under control of a T7 promoter. The rep and cap genes can be obtained from a variety of known sources. See, T. Shenk, J.
Virol., 61:3096-3101 (1987), which provides the AAV2 genome within the plasmid psub201; E. W. Lusby et al, J.
Virol., 41:518-526 (1982) and J. Smuda and B.J. Carter, Virologv, 184:310-318 (1991).
Similarly, the T7 promoter sequences J. Dunn and F.W. Studier, J. Mol. Biol., 166:477-535 (1983) may be obtained from a variety of commercial and academic sources. In a preferred embodiment, the vector further contains the sequence of untranslated region (UTR) of encephalomyocarditis (EMCV) downstream of the T7 WO 98/10088 PCTIUS97/15716 8 promoter. The inventors believe this sequence increases expression of the gene 5- to In addition, the vector also includes conventional regulatory elements necessary to drive expression of the rep/cap in a cell transfected with the vector. Such regulatory elements are known to those of skill in the art.
3. rAAV Cassette (Template) The third vector component contains a rAAV cassette containing a minigene flanked by AAV ITRs.
As discussed in more detail below, such a minigene contains a suitable transgene, a promoter, and other regulatory elements necessary for expression of the transgene.
The AAV sequences employed are preferably limited to the cis-acting 5' and 3' inverted terminal repeat (ITR) sequences [See, B. J. Carter, in "Handbook of Parvoviruses", ed., P. Tijsser, CRC Press, pp.155-168 (1990)]. Desirably, substantially the entire 143 bp sequences encoding the ITRs are used in the vectors. Some degree of minor modification of these sequences is expected to be permissible for this use.
The ability to modify these ITR sequences is within the skill of the art. See, texts such as Sambrook et al, "Molecular Cloning. A Laboratory Manual.", 2d edit., Cold Spring Harbor Laboratory, New York (1989).
Alternatively, it may be desirable to use functional fragments of the ITRs. Such fragments may be determined by one of skill in the art.
The AAV ITR sequences may be obtained from any known AAV, including presently identified human AAV types. Similarly, AAVs known to infect other animals may also be employed in the vector constructs of this invention. The selection of the AAV is not anticipated to limit the following invention. A variety of AAV WO 98/10088 PCT/US97/15716 9 strains, types 1-4, are available from the American Type Culture Collection or available by request from a variety of commercial and institutional sources. In the following exemplary embodiment an AAV-2 is used for convenience.
The 5' and 3' AAV ITR sequences flank a minigene which is made up of a selected transgene sequence and associated regulatory elements. The transgene sequence of the vector is a nucleic acid sequence heterologous to the AAV sequence, which encodes a polypeptide or protein of interest. The transgene is operatively linked to regulatory components in a manner which permits transgene transcription.
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. Such reporter sequences include without limitation an E. coli betagalactosidase (LacZ) cDNA, an alkaline phosphatase gene and a green fluorescent protein gene. These sequences, when associated with regulatory elements which drive their expression, provide signals detectable by conventional means, ultraviolet wavelength absorbance, visible color change, etc. A more preferred transgene sequence includes a therapeutic gene which expresses a desired gene product in a host cell. These therapeutic nucleic acid sequences typically encode products which may be administered to a patient in vivo or ex vivo to replace or correct an inherited or noninherited genetic defect or treat an epigenetic disorder or disease. The selection of the transgene sequence is not a limitation of this invention.
WO 98/10088 PCT/US97/15716 In addition to the major elements identified above, the minigene also includes conventional regulatory elements necessary to drive expression of the transgene in a cell transfected with the vector carrying the AAV cassette. Thus the minigene contains a selected promoter which is linked to the transgene and located within the minigene, between the AAV ITR sequences of the vector.
Selection of the promoter which mediates expression of the transgene is a routine matter and is not a limitation of the vector. Useful promoters include those which are discussed above in connection with the first vector component.
The minigene will also desirably contain heterologous nucleic acid sequences including sequences providing signals required for efficient polyadenylation of the transcript and introns with functional splice donor and acceptor sites. A common poly-A sequence which is employed in the exemplary vectors of this invention is that derived from the papovavirus SV-40. The poly-A sequence generally is inserted following the transgene sequences and before the 3' AAV ITR sequence. A common intron sequence is also derived from SV-40, and is referred to as the SV-40 T intron sequence. A minigene of the present invention may also contain such an intron, desirably located between the promoter/enhancer sequence and the transgene.
Selection of these and other common vector elements are conventional and many such sequences are available [see, Sambrook et al, and references cited therein].
The rAAV vector containing the AAV ITRs flanking the minigene may be carried on a plasmid backbone and used to transfect a selected host cell or may be flanked by viral sequences adenoviral sequences) which permit it to infect the selected host WO 98/10088 PCT/US97/15716 11 cell. Suitable Ad/AAV recombinant viruses may be produced in accordance with known techniques. See, e.g., WO 96/13598, WO 95/23867, and WO 95/06743, which are incorporated by reference herein.
B. Double Infection/Transfection A cell line which stably expresses T7 pol may be constructed, and then double transfected (or infected) with a vector containing T7/rep/cap and a vector containing a rAAV cassette, as illustrated in the following table (Inf infection and Txf transfection).
T7 rep/cap rAAV System A Inf Inf System B Inf Txf System C Txf Inf System D Txf Txf Alternatively, a cell line stably transfected with T7 rep/cap may be double transfected (infected) with a vector carrying T7 pol and a vector carrying the rAAV cassette, as illustrated in the following table.
T7 Pol rAAV System E Inf Inf System F Inf Txf System G Txf Inf System H Txf Txf In still another alternative, a cell line which contains a rescuable rAAV cassette may be double transfected (infected) with a vector containing T7 Pol and a vector containing T7/rep/cap, as illustrated in the following table.
T7 Pol T7 rep/cap System I Inf Inf System J Inf Txf System K Txf Inf System L Txf Txf WO 98/10088 PCT/US97/15716 12 The plasmid and viral vectors used in double transfection/infection steps are as described above in connection with the triple transfection and/or infection system.
A stable cell line of the invention can be produced by transfection of a desired cell, 293 cells or other packaging cell lines expressing required adenoviral genes, with a plasmid containing the desired gene, T7 Pol, using conventional techniques and selected via an accompanying resistant marker gene.
Depending upon whether inducible or constitutive expression is desired, an appropriate promoter may be selected. For example, if a host cell inducibly expressing T7 Pol is desired, the cell may be transfected with a plasmid containing T7 Pol under control of a metallothionein promoter. Alternatively, if a host cell constitutively expressing T7 Pol is desired, it may be inserted under control of a RSV or CMV promoter. Similar techniques may be used for providing a host cell containing the T7/rep/cap and a host cell containing a rescuable rAAV. The examples below describe production of stable cell lines. However, one of skill in the art could readily produce such cell lines using other conventional techniques. See, generally, Ausubel et al, Current Protocols in Molecular Biology (Wiley Interscience 1987).
C. Single Infection/Transfection A cell line which stably expresses two of the components of this system may be constructed, and then transfected (or infected) with a vector containing the remaining component of the system, as described above. For example, using the techniques described herein, a cell line is utilized which is stably transfected with the T7/rep/cap and a rescuable rAAV.
The cell line is then transfected or infected with a WO 98/10088 PCT/US97/15716 13 vector containing the T7 pol. As another example, the cell line is stably transfected with the T7 pol and a rescuable rAAV. The cell line is then transfected or infected with a vector containing the T7 rep/cap.
D. Cell Line Containing T7 Pol, rAAV and T7/rep/cap A cell line which stably expresses all three of the components of this system may be constructed and utilized in the method of the invention. Using known techniques, a suitable packaging cell line is constructed which contains the rAAV, the T7/rep/cap and the T7 pol.
In this embodiment, the T7 Pol is placed under the control of an inducible promoter. Suitable inducible promoters are known to those of skill in the art and are discussed herein. For example, T7 Pol may be placed under control of a metallothionein promoter. In this manner, expression of the T7 Pol, and thus the rep/cap, which are under control of the T7 promoter can be regulated and toxic effects to the cell avoided.
E. Production of Vectors and rAAV Assembly of the selected DNA sequences of the adenovirus, AAV and the reporter genes or therapeutic genes and other vector elements into the vectors described above utilize conventional techniques. Such techniques include cDNA cloning such as those described in texts [Sambrook et al, cited above], use of overlapping oligonucleotide sequences of the adenovirus or AAV genome, polymerase chain reaction, and any suitable method which provides the desired nucleotide sequence.
Whether using the three vector system, or stably infected cells, introduction of the vectors into the host cell is accomplished using known techniques.
Where appropriate, standard transfection and cotransfection techniques are employed, CaPO 4 WO 98/10088 PCT/US97/15716 14 transfection techniques using the complementation human embryonic kidney (HEK) 293 cell line (a human kidney cell line containing a functional adenovirus Ela gene which provides a transacting Ela protein). Other conventional methods employed in this invention include homologous recombination of the viral genomes, plaquing of viruses in agar overlay, methods of measuring signal generation, and the like.
Following infection/transfection, the host cell is then cultured under standard conditions, to enable production of the rAAV. See, F. L. Graham and L. Prevec, Methods Mol. Biol., 1:109-128 (1991).
Desirably, once the rAAV is identified using conventional techniques, it may be isolated using standard techniques and purified.
These examples illustrate the preferred methods of the invention. These examples are illustrative only and do not limit the scope of the invention.
Example 1 Construction of a T7 Pol Adenovirus Figure 1 provides a schematic of the construction of the recombinant adenovirus carrying the T7 polymerase.
The plasmid pMTT7N was obtained from Dr.
Michael Strauss Lieber et al, Nucl. Acids Res., 17:8485-8493 (1989)]. pMTT7N contains a N-terminal nuclear location signal of SV40 large T antigen fused to the T7 Pol gene (T7N Pol) which is linked to the polyadenylation sequence of SV40. Expression is driven by the inducible mouse metallothionein promoter.
The pMTT7N plasmid DNA was digested with BglII and PvuII restriction enzymes and the fragments separated on an agarose gel. The BglII/PvuII T7 Pol DNA fragment was ligated to the BglII/EcoRV cleaved vector pAd.CMV.link.l to form pAd.CMV.T7N. pAd.CMV.link.1 is a WO 98/10088 PCT/US97/15716 plasmid containing the adenoviral sequences 0 to 16 map units deleted of Ela and Elb into which a CMV promoter-polylinker cassette was cloned. This is described in X. Ye et al, J. Biol. Chem., 271:3639-3646 (1996).
In pAd.CMV.T7N, the expression unit of T7 Pol is directed by the CMV promoter. The promoter for the T7 Pol gene is linked to a PolyA tail as a cassette within the sequence of adenovirus 0-1 map unit (mu) and 9-16 mu.
The pAd.CMV.T7N is linearized by Nhe I digestion and cotransfected with Cla I linearized Addel327 backbone using Cellphate kit (Pharmacia). Approximately 1 week posttransfection, the T7 Pol adenovirus can be isolated from the plaques for further purification.
Example 2 Cell Lines Expressin T7 Pol A cell line stably expressing T7 Pol is established by co-transfection of plasmids pMTT7N and pMTCB6+ (which provides a selective marker) H. Choo et al, DNA, 5:529-538; Eur. J. Biochem., 174:417-424] into 293 cell at a ratio of 10:1 using calcium phosphate precipitation Graham and A. van der Eb, Virol., 52:456-467 (1973)]. Colony cloning is carried out by Geneticin selection at a concentration of 1 mg/ml. Each clone obtained is transfected with pT7 rep/cap plasmid [see, Example 3 below] and analyzed for its ability to induce the expression of Rep protein upon induction by supplementation with Zn++.
To establish a stable cell line that constitutively expresses the T7 Pol, the T7N Pol (obtained by BglII/PvuII digestion of pMTT7N, as described above) was subcloned downstream of RSV promoter at the cloning sites of BamHI and PvuII in the vector of pEBVhis [Invitrogen]. The resulting plasmid, designated pEBVhisT7N, was transfected into 293 cells and selected WO 98/10088 PCT/US97/15716 16 with Hygromycin at a concentration of 400 pg/ml. Each positive clone is analyzed for the presence of T7 Pol by its ability to produce expression of T7-LacZ or T7rep/cap in cells transfected with these plasmids.
Example 3 Production of T7 rep/cap Adenovirus The production of this recombinant adenoviral vector is illustrated schematically in Figs. 2 and 3.
A. Plasmid Construction The plasmid pTM1 Moss et al, Nature, 348:91-92 (1990)], designed for expressing genes under control of the T7 promoter/EMCV UTR (untranslated region of encephalomyocarditis), was used as the vector for expressing AAV rep/cap. The entire coding sequence of rep/cap was separated into two portions by the unique SacI site and subcloned into the pTMI plasmid as described below.
Because there is no appropriate restriction enzyme existing between the initiation site of rep and its natural promoter, p5, the left end of the rep sequence (N-rep) was first amplified by PCR. The sequence of the upper primer was SEQ ID NO:2: TATTTAAGCCCGAGTGAGCT (from position of 255 to 274) which introduced a nucleotide substitution A->T at position 274 (underlined). A SacI site was then generated to permit the cloning of N-rep into pTMl and in-frame expression of Rep protein from the EMCV UTR preferred initiation site (within the NcoI site). The PCR product (739 bp in length) was directly cloned into pCR2.1 vector (Invitrogen) and named pCR-N-rep.
The pTM-1 plasmid was digested with Sad and Stu I restriction enzymes and ligated with a 3.7 kb SacI/SnaBI fragment from psub201 [Samulski et al, J.
Virol., 61:3096-3101 (1987)] containing the right end of the AAV genome (without ITR sequence), the c- WO 98/10088 PCT/US97/15716 17 terminal portion of rep and full-length cap sequence.
This T7 promoter-driven rep/cap construct is named pT7-crep/cap.
The first 535 bp sequence of rep was removed from the pCR-N-Rep plasmid by SacI digestion and subcloned into pT7-C-rep/cap, which has similarly been digested with SacI and subjected to alkaline phosphatase treatment to prevent self-ligation of the vector. The final construct was named pT7 rep/cap which contains the full length coding sequence of rep/cap downstream of T7 promoter/EMCV UTR, followed by the T7 terminating sequence.
B. Production T7 rep/cap Adenovirus pAd.link is a construct similar to pAd.CMV.link, a plasmid containing the adenoviral sequences 0 to 16 map units deleted of Ela and Elb as described in the other adenovirus vectors into which a CMV promoter-polylinker cassette was cloned and described in X. Ye et al, J. Biol. Chem., 271:3639-3646 (1996).
However, pAd.link contains no CMV promoter or polyA tail sequence.
The entire region including the T7 promoter, EMCV UTR, rep/cap and T7 terminating sequence was excised from pT7 rep/cap by digestion with Clal and EagI, and then subcloned into the adenoviral sequences of pAd.link, which had previously been subjected to ClaI/SalI digestion, after filling in the sticky ends of EagI and SalI by Klenow polymerase. The resulting plasmid is designated pAd.T7 rep/cap.
The pAd.T7 rep/cap is co-transfected with the Clal linearized Ad.de1327 backbone DNA into 293 cell for the generation of T7 rep/cap adenovirus.
WO 98/10088 PCT/US97/15716 18 Example 4 Cell Line Expressing rep/caD A cell line stably transfected with pT7 rep/cap is established by transfection of pMTCB6+ into 293 cell at ratio of 10:1 and selected with Geneticin. Each clone is analyzed for the presence of rep protein by transfection with T7 Pol expressing plasmid.
Example 5 Production of Recombinant AAV Hybrid Vector Plasmid pAV.CMVLacZ serves as a template for rAAV to be replicated and packaged in the presence of AAV non-structural and capsid proteins.
Plasmid AV.CMVLacZ is a rAAV cassette in which rep and cap genes are replaced with a minigene expressing B-galactosidase from a CMV promoter. The linear arrangement of AV.CMVLacZ includes: the 5' AAV ITR (bp 1-173) obtained by PCR using pAV2 A. Laughlin et al, Gene, 23: 65-73 (1983)] as template [nucleotide numbers 365-538 of SEQ ID NO:1]; a CMV immediate early enhancer/promoter [Boshart et al, Cell, 41:521-530 (1985); nucleotide numbers 563-1157 of SEQ ID NO:1], an SV40 intron (nucleotide numbers 1178- 1179 of SEQ ID NO:1), E. coli beta-galactosidase cDNA (nucleotide numbers 1356 4827 of SEQ ID NO:1), an SV40 polyadenylation signal (a 237 BamHI-BclI restriction fragment containing the cleavage/poly-A signals from both the early and late transcription units; nucleotide numbers 4839 5037 of SEQ ID NO:1) and 3'AAV ITR, obtained from pAV2 as a SnaBI- BglII fragment (nucleotide numbers 5053 5221 of SEQ ID NO:1).
WO 98/10088 PCT/US97/15716 19 Where desired, the LacZ gene can be replaced with a desired therapeutic or other transgene for the purpose of generating new rAAV. See, Fig. 4. The sequence including CMV directed LacZ reporter cassette in between two AAV ITR sequences is excised from pAV.CMV.LacZ by PvuII digestion. This fragment is ligated with the EcoRV treated pAd.link to generate the plasmid pAd.AV.CMVLacZ. This plasmid is co-transfected with Clal linearized Addel327 backbone DNA to generate an adeno-rAAV hybrid virus.
ExamDle 6 Cell line containihn rescuable, integrated rAAV template tp293 cells are transfected/infected with pAV.CMVLacZ/rAAV Ad hybrid virus to generate cell line 15 that has incorporated rAAV, as determined by analysis of the genomic DNA by Southern blot. The clone is examined for the rescue of rAAV template by transfection/infection with rep/cap expressing constructs.
Numerous modifications and variations of the 20 present invention are included in the above-identified specification and are expected to be obvious to one of skill in the art. Such modifications and alterations to the processes of the present invention are believed to be encompassed in the scope of the claims appended hereto.
Throughout the specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
WO 98/10088 PCT/US97/15716 SEQUENCE LISTING GENERAL INFORMATION: APPLICANT: Trustees of the University of Pennsylvania Wilson, James M.
Chen, Nancie N.
(ii) TITLE OF INVENTION: An Inducible Method for Production of Recombinant Adeno-Associated Viruses Utilizing T7 Polymerase (iii) NUMBER OF SEQUENCES: 2 (iv) CORRESPONDENCE ADDRESS: ADDRESSEE: Howson and Howson STREET: Spring House Corporate Cntr, PO Box 457 CITY: Spring House STATE: Pennsylvania COUNTRY: USA ZIP: 19477 COMPUTER READABLE FORM: MEDIUM TYPE: Floppy disk COMPUTER: IBM PC compatible OPERATING SYSTEM: PC-DOS/MS-DOS SOFTWARE: PatentIn Release Version #1.30 (vi) CURRENT APPLICATION DATA: APPLICATION NUMBER: WO FILING DATE:
CLASSIFICATION:
(vii) PRIOR APPLICATION DATA: APPLICATION NUMBER: US 60/024,699 FILING DATE: 06-SEP-1996 (viii) ATTORNEY/AGENT INFORMATION: NAME: Kodroff, Cathy A.
REGISTRATION NUMBER: 33,980 REFERENCE/DOCKET NUMBER: GNVPN.022CIP1PCT (ix) TELECOMMUNICATION INFORMATION: TELEPHONE: 215-540-9200 TELEFAX: 215-540-5818 INFORMATION FOR SEQ ID NOil: SEQUENCE CHARACTERISTICS: LENGTH: 10398 base pairs TYPE: nucleic acid STRANDEDNESS: double TOPOLOGY: unknown (ii) MOLECULE TYPE: cDNA WO 98/10088 WO 9810088PCTIUS97/15716 21 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:
GAATTCGCTA
GGGGGTGGAG
GGCGGAAGTG
TGACGTTTTT
GGATGTTGTA
GAATAAGAGG
AGGGAGATCT
GGGCGACCTT
ACTCCATCAC
TTCGAGCTTG
CCGCCCAACG
ATAGGGACTT
GTACATCAAG
CCCGCCTGGC
TACGTATTAG
GGATAGCGGT
TTGTTTTGGC
ACGCAAATGG
AACCGTCAGA
GACCGATCCA
GTTAACTGGT
CAAATCAAAG
TTACTTCTGC
GAGCCTGCTA
TTTTCGTTGC
ATCCCGTCGT
TTGCAGCACA
CTTCCCAACA
AAGCGGTGCC
GCATCATCAA
TTTGTGACGT
TGATGTTGCA
GGTGTGCGCC
GTAAATTTGG
AAGTGAAATC
GCTGCGCGCT
TGGTCGCCCG
TAGGGGTTCC
CATGCCTGCA
ACCCCCGCCC
TCC&TTGACG
TGTATCATAT
ATTATGCCCA
TCATCGCTAT
TTGACTCACG
ACCAAAATCA
GCGGTAGGCG
TCGCCTGGAG
GCCTCCGGAC
AAGTTTAGTC
A&CTGCTCCT
TCTAAAAGCT
AAGCAAAAAA
CGGTCTGGGA
TTTACAACG7
*TCCCCCTTTC
*GTTGCGCAGC
GGAAAGCTGG
TAATATACCT
GGCGCGGGGC
AGTGTGGCGG
GGTGTACACA
GCGTAACCGA
TGAATAATTT
CGCTCGCTCA
GCCTCAGTGA
TTGTAGTTAA
GGTCGTTACA
ATTGACGTCA
TCAATGGGTG
GCCAAGTACG
GTACATGACC
TACCATGGTG
GGG;ATTTCCA
ACGGGACTTT
TGTACGGTGG
ACGCCATCCA
TCTAGAGGAT
TTTTTGTCTT
CAGTGGATGT
GCGGAATTGT
GAAGTCACCA
*GGCATTGGTC
*CGTGACTGGG
GCCAGCTGGC
CTGAATGGCG
TATTTTGGAT
GTGGGAACGG
AACACATGTA
GGAAGTGPICA
GTAAGATTTG
TGTGTTACTC
CTGAGGCCGC
GCGAGCGAGC
TGATTAACCC
TAACTTACGG
ATAATGACGT
GAGTATTTAC
CCCCCTATTG
TTATGGGACT
ATGCGGTTTT
AGTCTCCACC
CCAAAATGTC.
GAGGTCTATA
CGCTGTTTTG
CCGGTACTCG
TTATTTCAG
TGCCTTTACT
ACCCGCGGCC
TGTCGTTTAC
TGGACACCAG
AAAACCCTGG
GTAATAGCGA
AATGGCGCTT
TGAAGCCAAT ATGATAATGA
GGCGGGTGAC
AGCGACGGAT
ATTTTCGCGC
GCCATTTTCG
ATAGCGCGTA
CCGGGCAAAG
GCGCAGAGAG
GCCATGCTAC
TAAATGGCCC
ATGTTCCCAT
GGTAAACTGC
ACGTCAATGA
TTCCTACTTG
GGCAGTACAT
CCATTGACGT
GTAACAACTC
TAAGCAGAGC
ACCTCCATAG
AGGAACTGAA
TCCCGGATCC
TCTAGGCCTG
GCAATTCCCG
TTTGACCAAC
CAAGGAGCTG
CGTTACCCAA
AGAGGCCCGC
TGCCTGGTTT
GGCCGATACT
GTAGTAGTGT
GTGGCAAAAG
GGTTTTAGGC
CGGGAAAACT
ATATTTGTCT
CCCGGGCGTC
GGAGTGGCCA
TTATCTACAA
GCCTGGCTGA
AGTAACGCCA
CCACTTGGCA
CGGTAAATGG
GCAGTACATC
CAATGGGCGT
CAATGGGAGT
CGCCCCATTG
TCGTTTAGTG
AAGACACCGG
AAACCAGAAAL
GGTGGTGGTG
TACGGAAGTG
GGGATCGAAA
AAGAACGTGA
CTCAAGCGCG
CTTAATCGCC
ACCGATCGCC
CCGGCACCAG
GTCGTCGTCC
120 180 240 300 360 420 480 540 600 660 720 780 840 900 960 1020 1080 1140 1200 1260 1320 1380 1440 1500 1560 1620 1680 1740 1800 CTGGAGTGCG ATCTTCCTGA CCTCAAACTG GCAGATGCAC GGTTACGATG CGCCCATCTA CACCAACGTA ACCTATCCCA WO 98/10088 WO 9810088PCTIUS97/15716 22 TTACGGTCAA TCCGCCGTTT GTTCCCACGG AGARTCCGhC GGGTTGTTAC TCGCTCACAT
TTARTGTTGA
ACTCGGCGTT
TGCCGTCTGA
TGGTGCTGCG
GCATTTTCCG
TTGCCACTCG
GCGGCGAGTT
TCGCCAGCGG
ATCGCGTCAC
ATCTCTATCG
CCTGCGATGT
AGCCGTTGCT
TGYGATGAGCA
TGCGCTGTTC
TGTATGTGGT
CCGATGATCC
ATCGTAATCA
ATCACGACGC
AAGGCGGCGG
ATGAAGACCA
CTGGAGAGAC
GCGGTTTCGC
TCTGGGACTG
CTTACGGCGG
TCTTTGCCGA
TCCAGTTCCG
GCGATAACGA
AAGTGCCTCT
AGCCGGAGAG
CATGGTCAGA
TGAAAGCTGG
TCATCTGTGG
ATTTGACCTG
TTGGAGTGAC
TGACGTCTCG
CTTTAATGAT
GCGTGACTAC
CACCGCGCCT
ACTACGTCTG
TGCGGTGGTT
CGGTTTCCGC
GATTCGAGGC
GACGATGGTG
GCATTATCCG
GGATGAAGCC
GCGCTGGCTA
CCCGAGTGTG
GCTGTATCGC
AGCCGACACC
GCCCTTCCCG
GCGCCCGCTG
TAAATACTGG
GGTGGATCAG
TGATTTTGGC
CCGCACGCCG
TTTATCCGGG
GCTCCTGCAC
GGATGTCGC7
CGCCGGGCAA
AGCCGGGCAC
CTACAGGAAG
TGCAACGGGC
AGCGCATTTT
GGCAGTTATC
TTGCTGCATA
GATTTCAGCC
CTACGGGTAA
TTCGGCGGTG
AACGTCGAAA
GAACTGCACA
GAGGTGCGGA
GTTAACCGTC
CAGGPUTATCC
AACCATCCGC
AATATTGAAA
CCGGCGATGA
ATCATCTGGT
TGGATCAAAT
ACGGCCACCG
GCTGTGCCGA
ATCCTTTGCG
CAGGCGTTTC
TCGCTGATTA
GATACGCCGA
CATCCAGCGC
CAAACCATCG
TGGATGGTGG.
CCACAAGGT;
CTCTGGCTCZ
ATCAGCGCCI1
GCCAGACGCG
GCTGGGTCGG
TACGCGCCGG
TGGAAGATCA
AACCGACTAC
GCGCTGTACT
CAGTTTCTTT
AAATTATCGA
ACCCGAAACT
CCGCCGACGG
TTGAAAATGG
ACGAGCATCA
TGCTGATGAA
TGTGGTACAC
CCCACGGCAT
GCGAACGCGT
CGCTGGGGAA
CTGTCGATCC
ATATTATTTG
AATGGTCCAT
AATACGCCCA
GTCAGTATCC
AATATGATGA
ACGATCGCCA
TGACGGPJAGC
AAGTGACCAG-
r CGCTGGATGC
LAACAGTTGAI
kCAGTACGCGI1
?GGCAGCAGTC
AATTATTTTT
TTACGGCCAG
AGAAAACCGC
GGATATGTGG
ACAAATCAGC
GGAGGCTGAJ
ATGGCPLGGGT
TGAGCGTGGT
GTGGAGCGCC
CACGCTGATT
TCTGCTGCTG
TCCTCTGCAT
GCAGAACAAC
GCTGTGCGAC
GGTGCCAPLTG
AACGCGAATG
TGAATCAGGC
TTCCCGCCCG
CCCGATGTAC
CAAAAAATGG
CGCGATGGGT
CCGTTTACAG
MAACGGCAAC
GTTCTGTATG
AAAACACCAG
CGAATACCTG
ITAAGCCGCTG
TGAACTGCC7
AGTGCAACCG-
GCGTCTGGCC
GATGGCGTTA
GACAGTCGTT
CTCGCGGTGA
CGGATGAGCG
GATTTCCATG
GTTCAGATGT
GAAACGCAGG
GGTTATGCCG
GAAATCCCGA
GAAGCAGAP*G
CTGAACGGCA
GGTCAGGTCA
TTTAACGCCG
CGCTACGGCC
AATCGTCTGA
GTGCAGCGCG
CACGGCGCTA
GTGCAGTATG
GCGCGCGTGG
CTTTCGCTAC
AACAGTCTTG
GGCGGCTTCG
CCGTGGTCGG
'AACGGTCTGG
CAGCAGTTTT
TTCCGTCATA
GCAAGCGGTG
GAACTACCGC
AACGCGACCG
GAAAACCTCA
1860 1920 1980 2040 2100 2160 2220 2280 2340 2400 2460 2520 2580 2640 2700 2760 2820 2880 2940 3000 3060 3120 3180 3240 3300 3360 3420 3480 3540 3600 3660 WO 98/10088 WO 9810088PCT/US97/15716
GTGTGACGCT_
TTTGCATCGAi
AGATGTGGAT
CACCGCTGGA
TCGAACGCTG
CAGATACACT
AAACCTTATT
CCGTTGATGT
AGCTGGCGCA
CCGACCGCCT
CCCCGTACGT
GCCCACACCA
TGATGGAAAC
ACGGTTTCCA
TACAGCTGAG
CGGGCAGGCC
ACACAAACTT
ACTTCCCGTT
GTATTATTTT
TTTCTGACAA
ATTGATGAGT
ATTTGTGATG
AACAATTGCA
CCTCTAGAGT
AGTGATGGAG
AAAGGTCGCC
CAGATCTGGA
GGCGGTAAAC
GATCACTTGG
TGAGGTACTG
CCCCGCCGCG
GCTGGGTAAT
TGGCGATAA
TAACGACATT
GAAGGCGGCG4
TGCTGATGCG
TATCAGCCGG
TGAAGTGGCG
GGTAGCAGAG
TACTGCCGCC
CTTCCCGAGC
GTGGCGCGGC
CAGCCATCGC
TATGGGGATT
CGCCGGTCGC
ATGTCTGCCC
TTGGATGTTC
TTTCCCGATT
TGCCGCTATT
ACTCGGCCTC
TTGGACAAAC
CTATTGCTTT
TTCATTTTAT
CGAGTAGATA
TTGGCCACTC
CGACGCCCGG
AGGTGCTGAG
ATATTAGGAA
TGCTGGCCTG
AAATGTGTGG
rcccAcGccA kAGCGTTGGC PiARCAACTGC
GGCGTAAGTG
GGCCATTACC
GTGCTGATTA
AAAACCTACC
AGCGATACAC
CGGGTAAACT
TGTTTTGACC
GAAAACGGTC
GACTTCCAGT
CATCTGCTGC
GGTGGCGACG
TACCATTACC
GTATTTCGCG
GGTTTATTCT
TGGCTACATG
TCTCTGTTCT
GACTCTAGGC
CACAACTAGA
ATTTGTAACC
GTTTCAGGTT
AGTAGCATGG
CCTCTCTGCG
GCTTTGCCCG
GTACGATGAG
CCAGCCTGTG
CACCCGCGCI
GCGTGGCTTP
TCCCGCATCT
AATTTAACCG
TGACGCCGCT
AAGCGACCCG
AGGCCGAAGC
CGACCGCTCA
GGATTGATGG
CGCATCCGGC
GGCTCGGATT
GCTGGGATCT
TGCGCTGCGG
TCAACATCAG
ACGCGGAAGA
ACTCCTGGAG
AGTTGGTCTG
TAAGGAAATC
TTTTCTTTTA
ACATCAACCA
CGCTATTATT
GGCCGCGGGG
ATGCAGTGAA
ATTATAAGCT
CAGGGGGP&GG
CGGGTTAATC
CGCTCGCTCG
GGCGGCCTCA
ACCCGCACCA
ATGCTGGATG
GAGTTTGGC!I
AGGGTGGGAP
GACCACCAGC
CCAGTCAGGC
GCGCGATCAG
CATTGACCCT
AGCGTTGTTG
CGCGTGGCAG
TAGTGGTCAA
GCGGATTGGC
AGGGCCGCAA
GCCATTGTCA
GACGCGCGAA
CCGCTACAGT
AGGCACATGG
CCCGTCAGTA
GTGTCAAAAA
CATTATGTAC
CTTTTTTATC
TATCAGCAAA
CCAACCGCTG
ATCCAGACAT
AAAAATGCTT
GCAATAAACA
TGTGGGAGGT
ATTAACTACA
CTCACTGAGG
GTGAGCQAGC
GGTGCAGACC
TGACCGAGGA
CTAGCGATGA
LAGAATATATA
GAAATGGATT
TTTCTTTCAC
TTCACCCOTG
AACGCCTGGG
CAGTGCACGG
CATCAGGGGA
ATGGCGATTA
CTGAACTGCC
GAAPJACTATC
GACATGTATA
TTGAATTATG
CAACAGCAAC
CTGAATATCG
TCGGCGGAAT
TAATAATAAC
TATTTAAAAA
ATGGGAGCCT
AGTGATACGG
TTTGGTCTGC
GATAAGATAC
TATTTGTGAA
AGTTAACAALC
TTTTTCGGAT
AGGAACCCCT
CCGGGCGACC
GAGCGCGCAG
CTGCGAGTGT
GCTGAGGCCC
AGATACAGAT
.AGGTGGGGGT
3720 3780 3840 3900 3960 4020 4080 4140 4200 4260 4320 4380 4440 4500 4560 4620 4680 4740 4800 4860 4920 4980 5040 5100 5160 5220 5280 5340 5400 5460 5520 CTTATGTAGT TTTGTATCTG TTTTGCAGCA GCCGCCGCCG CCATGAGCAC CAACTCGTTT wo 98/10088 WO 9810988PCT1US97/15716
GATGGAAGCA
CAGAATGTGA
TTGACCTACG
GCCGCTGCAG
AGCAGTGCAG
TTGGATTCTT
CAGGTTTCTG
CCAGACTCTG
CGCGCGCGGT
AGGACGTGGT
TGGAGGTAGC
TAGCAGGAGC
GGCAGGCCCT
GATATGAGAT
CGGGGATTCA
TCATGT.AGCT
TTTTCCATGC
ATATTTCTGG
TTTACAAAGC
GCGTAGTTAC
TCTACCTGCG
AGCAGGTTCC
ACCGGGTGCA
ACTTCGTTAA
TCGCCGCCCA
TCCGCCGTAG
GTCACCTGC7
TTTCGCTGTA
GGCGCAGGG7
CGCTGGCCAC-
TTGTGAGCTC
TGGGCTCCAG
AGACCGTGTC
CCACCGCCCG
CTTCCCGTTC
TGACCCGGGA
CCCTGAAGGC
TTTGGATTTG
AGGCCCGGGA
AI4AGGTGACT
ACCACTGCAG
GCTGGGCGTG
TGGTGTAAGT
GCATCTTGGA
TGTTGTGCAG
TAGAAGGAAA
ATTCGTCCAT
GATCACTAAC
GCGGGCGGAG
CCTCACAGAT
GGGCGATGAA
TGAGCAGCTG
ACTGGTAGTT
GCATGTCCC7
*GCGATAGCAG
*GCATGCTTTI
CTACGGCATC
CGGCAGTAGIJ
CCTCGTCAGC
GGTGCGCTTC
ATATTTGACA
CATTGATGGT
TGGAAICGCCG
CGGGATTGTG
ATCCGCCCGC
ACTTAATGTC
TTCCTCCCCT
GATCAAGCAA
CCAGCGGTCT
CTGGATGTTC
AGCTTCATGC
GTGCCTAAAA
GTTTACAAAG
CTGTATTTTT
AACCACCAGC
TGCGTGGAAG
AATGATGGCA
GTCATAGTTG
GGTGCCAGAC
TTGCATTTCC
GAAAACGGT7
CGACTTACCG
AALGAGAGCTC
GACTCGCATC
TTCTTGCAAC
GAGCGTTTGI
TCGATCCAG(
CGGTGCTCGI
GTAGTCTGG(
AGGCTGGTC(
ACGCGCATGC
CGCCCCGTCC
TTGGAGACTG
ACTGACTTTG
GATGACAAGT
GTTTCTCAGC
CCCAATGCGG
GTGTCTTGCT
CGGTCGTTGA
AGATACATG
TGCGGGOTGG
ATGTCTTTCA
CGGTTAAGCT
AGGTTGGCTA
ACAGTGTATC
AACTTGGAGA
ATGGGCCCAC
TGTTCCAGGA
TGCGGTATAA
CACGCTTTGA
TCCGGGGTAG
CAGCCGGTGG
ICAGCTGCCGT
TTTTCCCTG1A GAAGCAAAG7 iCCPAGCAGT~I
IATATCTCCTC
rCCAGACGGGC
TCACGGTGAI
STGCTGGTGC']
CCCCATGGGC
TGCCCGCAAA
CAGCCTCCGC
CTTTCCTGAG
TGACGGCTCT
AGCTGTTGGA
TTTAAAACAT
GTCTTTATTT
GGGTCCTGTG
GCATAAGCCC
TGTTGTAGAT
GTAGCAAGCT
GGGATGGGTG
TGTTCCCAGC
CGGTGCACTT
CGCCCTTGTG
GGGCGGCGGC
TGAGATCGTC
TGGTTCCATC
GTTCAGATGG
GGGAGATCAG
GCCCGTAAAT
CATCCCTGAG
CCAPAATCCGC
TTTTCAACGG
CCAGGCGGTC
GTTTCGCGGG
CAGGGTCATG
GGGGTGCGCT
GAAGCGCTGC
CGGGGTGCGT
CTCTACTACC
CGCCGCTTCA
CCCGCTTGCA
TTTGGCACAA
TCTGCGCCAG
AAATAAAAAA
AGGGGTTTTG
TATTTTTTCC
GTCTCTGGGG
GATCCAGTCG
GATTGCCAGG
CATACGTGGG
CATATCCCTC
GGGAAATTTG
ACCTCCAAGA
CTGGGCGAAG
ATAGGCCATT
CGGCCCAGGG
GGGGATCATG
CTGGGAAGAA
CACACCTATT
CAGGGGGGCC
CAGAAGGCGC
TTTGAGACCG
CCACAGCTCG
TTGGGGCGGC
TCTTTCCACG
CCGGGCTGCG
CGGTCTTCGC
5580 5640 5700 5760 5820 5880 5940 6000 6060 6120 6180 6240 6300 6360 6420 6480 6540 6600 6660 6720 6780 6840 6900 6960 7020 7080 7140 7200 7260 7320 7380 CCTGCGCGTC GGCCAGGTAG CATTTGACCA TGGTGTCATA GTCCAGCCCC TCCGCGGCGT 1. WO 98/10088 WO 9810088PCTIUS97/15716
GGCCCTTGGC
TGAGGGCGTA
AGGCCCCGCA
AAACCAGGTT
GTCCACGCTC
CCTCGACCGA
ATGACTATCG
CCGGCAGCGC
GGCCTGTCGC
CCCGCCACCA
CTGGGCTACG
CTTCTCGCTT
GATGACGACC
ATCACTGGAC
TTGGCATGGA
GCATGGAGCC
CCACTCCAAG
CTTGGCAGAA
GCAGCGTTGG
TAGGCTGGCG
GAAGCGACTO
TCCGTGTTTC
TCTGCATCGC
CTTTCTCAAT
GGCTGTGTGC
CTTGAGTCCA
ATTAGCAGAG
GGCTACACTA
AAAAGAGTTG
GTTTGCAAGC
TCTACGGGGT
GCGCAGCTTG
GAGCTTGGGC
GACGGTCTCG
TCCCCCATGC
GGTGACGAAA
TGCCCTTGAG
TCGCCGCACT
TCTGGGTCAT
TTGCGGTATT
AACGTTTCGG
TCTTGCTGGC
CCGGCGGCAT
ATCAGGGACA
CGCTGATCGT
TTGTAGGCGC
GGGCCACCTC
AATTGGAGCC
CATATCCATC
GTCCTGGCCA
GGGTTGCCTT
CTGCTGCAAA
GTAAAGTCTG
AGGATGCTGC
GCTCACGCTG
ACGAACCCCC
ACCCGGTAAiG
CGAGGTATGT
GAAGGACAGT
GTAGCTCTTG
AGCAGATTAC
CTGACGCTCA
CCCTTGGAGG
GCGAGAAATA
CATTCCACGA
TTTTTGATGC
AGGCTGTCCG
AGCCTTCAAC
TATGACTGTC
TTTCGGCGAG
CGGAATCTTG
CGAGAAGCAG
GTTCGCGACG
CGGGATGCCC
GCTTCAAGGA
CACGGCGATT
CGCCCTATAC
GACCTGAATG
AATCAATTCT
GCGTCCGCCA
CGGGTGCGCA
ACTGGTTAGC
ACGTCTGCGA
GAAACGCGGA
TGGCTACCCT
TAGGTATCTC
CGTTCAGCCC
ACACGACTTA
AGGCGGTGCT
ATTTGGTATC
ATCCGGCAAA
GCGCAGAAAA
GTGGAACGAA
AGGCGCCGCA
CCGATTCCGG
GCCAGGTGAG
GTTTCTTACC
TGTCCCCGTA
CCAGTCAGCT
TTCTTTATCA
GACCGCTTTC
CACGCCCTCG
GCCATTATCG
CGAGGCTGGA
GCGTTGCAGG
TCGCTCGCGG
TATGCCGCCT
CTTGTCTGCC
GAAGCCGGCG
TGCGGAGAAC
TCTCCAGCAG
TGATCGTGCT
AGAATGAATC
CCTGAGCAAC
AGTCAGCGCC
GTGGAACACC
AGTTCG4GTGT
GACCGCTGCG
TCGCCACTGG
ACAGAGTTCT
TGCGCTCTGC
CAAACCFICCG
AAAGGATCTC
AACTCACGTT
CGAGGGGCAG
GGAGTAGGCA
CTCTGGCCGT
TCTGGTTTCC
TACAGACTTG
CCTTCCGGTG
TGCAACTCGT
GCTG4GAGCGC
CTCAAGCCTT
CCGGCATGGC
TGGCCTTCCC
CCATGCTGTC
CTCTTPLCCAG
CGGCGAGCAC
TCCCCGCGTT
GCACCTCGCT
TGTGAATGCG
CCGCACGCGG
CCTGTCGTTG
ACCGATACGC
AACATGAATG
CTGCACCATT
TACATCTGTA
AGGTCGTTCG
CCTTATCCGG
CAGCAGCCAC
TGAAGTGGTG
TGAAGCCAGT
CTG4GTAGCGG
AAGAAGATCC
AAGGGATTTT
TGCAGACTTT
TCCGCGCCGC
TCGGGGTCAA
ATGAGCCGGT
AGAGGCCTGT
GGCGCGGGGC
AGGACAGGTG
GACGATGATC
CGTCACTGOT
GGCCGACGCG
CATTATGATT
CAGGCAGGTA
CCTAACTTCG
ATGGAACGGG
GCGTCGCGGT
AACGGATTCA
CAAACCAACC
CGCATCTCGG
AGGACCCGGC
GAGCGAACGT
GTCTTCGGTT
ATGTTCCGGA
TTAACGAAGC
CTCCAAGCTG
TAACTATCGT
TGGTAACAGG
GCCTAACTAC
TACCTTCGGA
TGGTTTTTTT
TTTGATCTTT
GGTCATGAGA
7440 7500 7560 7620 7680 7740 7800 7860 7920 7980 8040 8100 8160 8220 8280 8340 8400 8460 8520 8580 8640 8700 8760 8820 8880 8940 9000 9060 9120 9180 9240 WO 98/10088 WO 9810088PCTIUS97/15716
TTATCAAAAA
TAAAGTATAT
ATCTCAGCGA
ACTACGATAC
CGCTCACCGG
AGTGGTCCTG
GTAAGTAGTT
GTGTCACGCT
GTTACATGAT
GTCAGAAGTA
CTTACTGTCA
TTCTGAGAAT
ACCGCGCCAC
AAACTCTCAA
AACTGATCTT
CAAAATGCCG
CTTTTTCAAT
GAATGTATTT
CCTGACGTCT
AGGCCCTTTC
GGATCTTCAC
ATGAGTAAAC
TCTGTCTATT
GGGAGGGCTT
CTCCAGATTT
CAACTTTATC
CGCCAGTTAA
CGTCGTTTGG
CCCCCATGTT
AGTTGGCCGC
TGCCATCCGT
AGTGTATGCG
ATAGCAGAAC
GGATCTTACC
CAGCATCTTT
CAAAAAAGGG
ATTATTGAAG
AGAAAAATAA
AAGAAACCAT
GTCTTCAAi
CTAGATCCTT
TTGGTCTGAC
TCGTTCATCC
ACCATCTGGC
ATCAGCAATA
CGCCTCCATC
TAGTTTGCGC
TATGGCTTCA
GTGCAAAAAA
AGTGTTATCA
AAGATGCTTT
GCGACCGAGT
TTTAAAAGTG
GCTGTTGAGA
TACTTTCACC
AATAAGGGCG
CATTTATCAG
ACAAATAGGG
TATTATCATG
TTAAATTAAA
AGTTACCAMT
ATAGTTGCCT
CCCAGTGCTG
AACCAGCCLG
CAGTCTATTA
AACGTTGTTG
TTCAGCTCCG,
GCGGTTAGCT
CTCATGGTTA
TCTGTGACTG
TGCTCTTGCC
CTCATCATTG
TCCAGTTCGA
AGCGTTTCTG
ACACGGAAAT
GGTTATTGTC
GTTCCGCGCA
ACATTAACCT
AATGAAGTTT
GCTTAATCAG
GACTCCCCGT
CAATGATACC
CCGGAAGGGC
ATTGTTGCCG
CCATTGCTGC
GTTCCCAACG
CCTTCGGTCC
TGGCAGCACT
GTGAGTACTC
CGGCGTCAAC
GAAAACGTTC
TGTAACCCAC
GGTGAGCAAA
GTTGAATACT
TCATGAGCGG
CATTTCCCCG
ATAAAAATAG
TAAATCAATC!
TGAGGCACCT
CGTGTAGATA
GCGAGACCCA
CGAGCGCAGA
GGAAGCTAGA
AGGCATCGTG
ATCAAGGCGA
TCCGATCGTT
GCATAATTCT
AACCAAGTCA
ACGGGATAAT
TTCGGGGCGA
TCGTGCACCC
AACAGGAAGG
CATACTCTTC
ATACATATTT
AAAAGTGCCA
GCGTATCACG
9300 9360 9420 9480 9540 9600 9660 9720 9780 9840 9900 9960 10020 10080 10140 10200 10260 10320 10380 10398 INFORMATION FOR SEQ ID NOs2: SEQUENCE CHARACTERISTICSt LENGTH: 20 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: unknown (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2: TATTTAAGCC CGAGTGAGCT

Claims (22)

1. A method for production of recombinant adeno-associated virus (AAV) comprising the steps of: introducing into a selected host cell a first vector comprising T7 polymerase under control of sequences which drive expression thereof, a second vector comprising AAV rep and cap genes under control of T7 promoter sequences which drive expression of rep and cap; and a third vector comprising from 5' to a cassette consisting essentially of a 5' AAV inverted terminal repeat (ITR), a selected minigene, and a 3' AAV ITR; culturing the host cell under conditions which permit replication and packaging of recombinant AAV; and recovering the recombinant AAV.
2. The method according to claim 1 wherein at least one of the vectors is an adenovirus and the host cell is a 293 cell.
3. The method according to claim 1 wherein the first vector is a recombinant adenovirus.
4. The method according to claim 1 wherein the second vector is a recombinant adenovirus. The method according to claim 1 wherein the third vector further comprises adenoviral sequences flanking the cassette. WO 98/10088 PCT/US97/15716 28
6. The method according to any of claims 1 to wherein the minigene contains a transgene which is a marker gene.
7. The method according to claim 6 wherein the minigene contains a transgene which is a therapeutic gene.
8. A method for production of recombinant adeno-associated virus (AAV) comprising the steps of: providing a host cell which expresses T7 polymerase; introducing into the host cell a first vector which comprises AAV rep and cap genes under control of T7 promoter sequences; introducing into the host cell a second vector comprising a cassette consisting essentially of 5' AAV inverse terminal repeat (ITR), a selected minigene, and 3' AAV ITR; and culturing the host cell under conditions which permit replication and packaging of a recombinant AAV.
9. The method according to claim 8 wherein step comprises the step of transfecting the host cell with a vector comprising the T7 promoter and the AAV rep and cap genes. The method according to claim 8 wherein step comprises the step of infecting the host cell with a recombinant adenovirus comprising the T7 promoter sequences, and the AAV rep and cap genes. WO 98/10088 PCTIUS97/15716 29
11. The method according to claim 8 wherein step comprises transfecting the host cell with a vector comprising the cassette.
12. The method according to claim 8 wherein step comprises infecting the host cell with a recombinant adenovirus comprising the cassette flanked by adenovirus sequences.
13. A method for production of recombinant adeno-associated virus (AAV) comprising the steps of: providing a host cell stably I transfected with AAV rep and cap genes under control of T7 promoter sequences; introducing into the host cell a vector comprising T7 polymerase; introducing into the host cell a vector comprising a cassette consisting essentially of a 5' AAV inverse terminal repeat (ITR), a selected minigene, and a 3' AAV ITR; and culturing the host cell under conditions which permit replication and packaging of a recombinant AAV.
14. The method according to claim 13 wherein step comprises the step of transfecting the host cell with a vector comprising the T7 polymerase gene. The method according to claim 13 wherein step comprises the step of infecting the host cell with a recombinant adenovirus comprising the T7 polymerase gene under control of regulatory sequences SRA4'controlling expression thereof. lco uj WO 98/10088 PCT/US97/15716
16. The method according to claim 13 wherein step comprises the step of transfecting the host cell with a vector comprising the cassette.
17. The method according to claim 13 wherein step comprises the step of infecting the host cell with a recombinant adenovirus comprising the cassette flanked by adenovirus sequences.
18. A method for production of recombinant adeno-associated virus (AAV) comprising the steps of: providing a host cell comprising a cassette consisting essentially of 5' AAV inverse terminal repeats (ITR), a selected minigene, and a 3' AAV ITR; introducing into the host cell a vector comprising AAV rep and cap genes under control of T7 promoter sequences; introducing into the host cell a vector comprising the T7 polymerase; and culturing the host cell under conditions which permit replication and packaging of a recombinant AAV.
19. The method according to claim 18 wherein step comprises the step of transfecting the host cell with a plasmid vector. The method according to claim 18 wherein step comprises the step of infecting the host cell with a recombinant adenoviral vector. WO 98/10088 PCT/US97/15716 31
21. The method according to claim 18 wherein step comprises the step of transfecting the host cell with a plasmid vector containing the T7 polymerase under control of regulatory sequences which direct expression thereof.
22. The method according to claim 18 wherein step comprises the step of infecting the host cell with a recombinant adenovirus comprising the T7 polymerase under control of regulatory sequences which direct expression thereof.
23. A method for production of recombinant adeno-associated virus (AAV) comprising the steps of: providing a host cell stably transfected with a cassette consisting essentially of AAV inverse terminal repeats (ITR), a selected minigene, and a 3' AAV ITR and a plasmid comprising AAV rep and cap genes under control of T7 promoter sequences; introducing into the host cell a vector comprising the T7 polymerase; and culturing the host cell under conditions which permit replication and packaging of a recombinant AAV.
24. A method for production of recombinant adeno-associated virus (AAV) comprising the steps of; providing a host cell stably transfected with a cassette consisting essentially of AAV inverse terminal repeats (ITR), a selected minigene, and a 3' AAV ITR and a plasmid comprising T7 polymerase; introducing into the host cell a vector comprising AAV rep and cap genes under control of T7 promoter sequences; and WO 98/10088 PCTIUS97115716 32 culturing the host cell under conditions which permit replication and packaging of a recombinant AAV. A method for production of recombinant adeno-associated virus (AAV) comprising the steps of: providing a host cell stably transfected with a cassette consisting essentially of 5' AAV inverse terminal repeats (ITR), a selected minigene, and a 3' AAV ITR; su, r (ii) a plasmid comprising T7 polymerase under control of sequences which regulate expression thereof, said sequences comprising an .inducible promoter; and (iii) a plasmid AAV rep and cap genes under control of T7 promoter sequences; and inducing expression of said T7 promoter.
26. A recombinant adeno-associated virus (AAV) produced according to the method of any one of claims 1-25.
27. Methods for the production of a recombinant adeno-associated virus (AAV) or a recombinant adeno-associated virus produced therefrom substantially as herein described with reference to the examples and accompanying figures. DATED this sixteenth day of May, 2000. Si 1tRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA SIts Patent Attorneys *AVIES COLLISON CAVE
AU41833/97A 1996-09-06 1997-09-04 An inducible method for production of recombinant adeno-associated viruses utilizing T7 polymerase Ceased AU722624B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US2469996P 1996-09-06 1996-09-06
US60/024699 1996-09-06
PCT/US1997/015716 WO1998010088A1 (en) 1996-09-06 1997-09-04 An inducible method for production of recombinant adeno-associated viruses utilizing t7 polymerase

Publications (2)

Publication Number Publication Date
AU4183397A AU4183397A (en) 1998-03-26
AU722624B2 true AU722624B2 (en) 2000-08-10

Family

ID=21821933

Family Applications (1)

Application Number Title Priority Date Filing Date
AU41833/97A Ceased AU722624B2 (en) 1996-09-06 1997-09-04 An inducible method for production of recombinant adeno-associated viruses utilizing T7 polymerase

Country Status (6)

Country Link
EP (1) EP0931158A1 (en)
JP (1) JP2001500015A (en)
AU (1) AU722624B2 (en)
CA (1) CA2264482A1 (en)
IL (1) IL128780A0 (en)
WO (1) WO1998010088A1 (en)

Families Citing this family (216)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6924128B2 (en) 1994-12-06 2005-08-02 Targeted Genetics Corporation Packaging cell lines for generation of high titers of recombinant AAV vectors
US6989264B2 (en) 1997-09-05 2006-01-24 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US6953690B1 (en) 1998-03-20 2005-10-11 The Trustees Of The University Of Pennsylvania Compositions and methods for helper-free production of recombinant adeno-associated viruses
CA2342283C (en) * 1998-09-11 2004-05-25 The Regents Of The University Of California Recombinant adenovirus for tissue specific expression in heart
US6759237B1 (en) 1998-11-05 2004-07-06 The Trustees Of The University Of Pennsylvania Adeno-associated virus serotype 1 nucleic acid sequences, vectors and host cells containing same
JP4693244B2 (en) 1999-03-18 2011-06-01 ザ・トラステイーズ・オブ・ザ・ユニバーシテイ・オブ・ペンシルベニア Compositions and methods for helperless production of recombinant adeno-associated virus
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
US6893865B1 (en) 1999-04-28 2005-05-17 Targeted Genetics Corporation Methods, compositions, and cells for encapsidating recombinant vectors in AAV particles
US6793926B1 (en) 1999-05-27 2004-09-21 Genovo, Inc. Methods for production of a recombinant adeno-associated virus
US7115391B1 (en) 1999-10-01 2006-10-03 Genovo, Inc. Production of recombinant AAV using adenovirus comprising AAV rep/cap genes
DE60209193T2 (en) 2001-11-13 2006-09-28 Trustees Of The University Of Pennsylvania Method for identifying adeno-associated virus (AAV) sequences and kit for carrying out the method
HU230364B1 (en) 2001-11-21 2016-03-29 The Trustees Of The University Of Pennsylvania Simian adeniovirus nucleic acid and amino acid sequences, vectors containing same, and methods of use
EP1944043A1 (en) 2001-11-21 2008-07-16 The Trustees of the University of Pennsylvania Simian adenovirus nucleic acid and amino acid sequences, vectors containing same, and methods of use
DK2359869T3 (en) 2001-12-17 2019-04-15 Univ Pennsylvania Sequences of adeno-associated virus (AAV) serotype 8, vectors containing these, and uses thereof
EP2573170B1 (en) 2001-12-17 2017-12-20 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) serotype 9 sequences, vectors containing same, and uses therefor
IL163988A0 (en) 2002-03-15 2005-12-18 Wyeth Corp Mutants of the p4 protein of nontypable haemophilus influenzae with reduced enzymatic activity
DK1944318T3 (en) 2003-07-21 2011-06-14 Transgene Sa Multifunctional cytokines
US20070073264A1 (en) 2003-09-26 2007-03-29 The Trustees Of The University Of Pennsylvania Methods, compositions and apparatus for delivering heterologous molecules to cells
PT2292780T (en) 2003-09-30 2017-11-28 Univ Pennsylvania Adeno-associated virus (aav) clades, sequences, vectors containing same, and uses thereof
TW200613554A (en) 2004-06-17 2006-05-01 Wyeth Corp Plasmid having three complete transcriptional units and immunogenic compositions for inducing an immune response to HIV
WO2006039218A2 (en) 2004-09-30 2006-04-13 The Trustees Of The University Of Pennsylvania Perfusion circuit and use therein in targeted delivery of macromolecules
US7517870B2 (en) 2004-12-03 2009-04-14 Fondazione Telethon Use of compounds that interfere with the hedgehog signaling pathway for the manufacture of a medicament for preventing, inhibiting, and/or reversing ocular diseases related with ocular neovascularization
DE23193601T1 (en) 2005-04-07 2024-02-15 The Trustees Of The University Of Pennsylvania METHOD FOR INCREASE THE FUNCTION OF AN AAV VECTOR
AU2007277081B2 (en) 2006-07-27 2014-02-06 Wyeth Llc High-cell density fed-batch fermentation process for producing recombinant protein
CA2707029A1 (en) 2007-11-28 2009-08-27 Soumitra Roy Simian subfamily c adenoviruses sadv-40, -31, and -34 and uses thereof
BRPI0819783A2 (en) 2007-11-28 2015-06-23 Univ Pennsylvania Simian subfamily and adenovirus sadv-39, -25.2, -26, -30, -37 and -38 and uses thereof.
CN102016011B (en) 2008-03-04 2013-12-11 宾夕法尼亚大学托管会 Simian adenoviruses SADV-36,-42.1, -42.2, and -44 and uses thereof
US8940290B2 (en) 2008-10-31 2015-01-27 The Trustees Of The University Of Pennsylvania Simian adenoviruses SAdV-43, -45, -46, -47, -48, -49, and -50 and uses thereof
ES2724122T3 (en) 2009-04-30 2019-09-06 Univ Pennsylvania Compositions for directing conductive airway cells comprising adeno-associated virus constructs
CN102575232B (en) 2009-05-29 2015-07-22 宾夕法尼亚大学托管会 Simian adenovirus 41 and uses thereof
US20120309011A1 (en) * 2009-11-02 2012-12-06 Salk Institute For Biological Studies Targeting of modifying enzymes for protein evolution
CA3066596A1 (en) 2010-04-23 2011-10-27 University Of Massachusetts Cns targeting aav vectors and methods of use thereof
JP5719443B2 (en) 2010-09-26 2015-05-20 ダ・ユー・エンタープライジズ、エルエルシー Recombinant macromolecule production method
AU2011332025B2 (en) 2010-11-23 2015-06-25 The Trustees Of The University Of Pennsylvania Subfamily E simian adenoviruses A1321, A1325, A1295, A1309 and A1322 and uses thereof
US9109219B2 (en) 2010-12-09 2015-08-18 Institut Pasteur MGMT-based method for obtaining high yields of recombinant protein expression
SG10202007803QA (en) 2011-02-17 2020-09-29 Univ Pennsylvania Compositions and methods for altering tissue specificity and improving aav9-mediated gene transfer
EP2710130B1 (en) 2011-05-19 2022-06-29 Fundación Pública Andaluza Progreso Y Salud Highly inducible dual-promoter lentiviral tet-on system
EP2760463B1 (en) 2011-09-20 2018-11-21 The University of North Carolina At Chapel Hill Regulation of sodium channels by plunc proteins
US10640785B2 (en) 2011-11-22 2020-05-05 The Children's Hospital Of Philadelphia Virus vectors for highly efficient transgene delivery
KR102007061B1 (en) 2011-12-09 2019-08-02 앵스띠뛰 파스퇴르 Multiplex immuno screening assay
SG11201406776TA (en) 2012-04-18 2015-03-30 Philadelphia Children Hospital Composition and methods for highly efficient gene transfer using aav capsid variants
CN105473723A (en) 2012-05-18 2016-04-06 宾夕法尼亚大学托管会 Subfamily E simian adenoviruses A1302, A1320, A1331 and A1337 and uses thereof
EP3587455A1 (en) 2012-10-23 2020-01-01 Emory University Gm-csf and il-4 conjugates, compositions, and methods related thereto
WO2014068072A1 (en) 2012-10-31 2014-05-08 Institut Gustave-Roussy Identification, assessment and therapy of essential thrombocythemia with resistance to jak2 inhibitors
WO2014071182A1 (en) 2012-11-01 2014-05-08 Massachusetts Institute Of Technology Directed evolution of synthetic gene cluster
EP2917341B1 (en) 2012-11-08 2017-09-27 Centre National de la Recherche Scientifique (CNRS) New phosphodeoxyribosyltransferase mutant enzymes and uses thereof
CN105026554B (en) 2013-03-15 2020-08-11 宾夕法尼亚大学托管会 Compositions and methods for treating MPS1
MX2016000345A (en) 2013-07-12 2016-06-21 Philadelphia Children Hospital Aav vector and assay for anti-aav (adeno-associated virus) neutralizing antibodies.
KR20220041963A (en) 2013-07-22 2022-04-01 더 칠드런스 호스피탈 오브 필라델피아 Variant aav and compositions, methods and uses for gene transfer to cells, organs and tissues
LT3116900T (en) 2014-03-09 2020-12-10 The Trustees Of The University Of Pennsylvania Compositions useful in treatment of ornithine transcarbamylase (otc) deficiency
WO2015164786A1 (en) 2014-04-25 2015-10-29 University Of Massachusetts Recombinant aav vectors useful for reducing immunity against transgene products
AU2015252797C1 (en) 2014-05-02 2021-04-22 Genzyme Corporation AAV vectors for retinal and CNS gene therapy
EP3151866B1 (en) 2014-06-09 2023-03-08 Voyager Therapeutics, Inc. Chimeric capsids
CN107208069A (en) 2014-07-16 2017-09-26 特兰斯吉恩股份有限公司 Oncolytic virus for expressing immunologic test point regulatory factor
JP6842410B2 (en) 2014-10-03 2021-03-17 ユニバーシティ オブ マサチューセッツ AAV vector identified by a novel high efficiency library
RU2020140209A (en) 2014-10-21 2021-01-25 Юниверсити Оф Массачусетс RECOMBINANT AAV OPTIONS AND THEIR APPLICATIONS
EP3215191A4 (en) 2014-11-05 2018-08-01 Voyager Therapeutics, Inc. Aadc polynucleotides for the treatment of parkinson's disease
WO2016077689A1 (en) 2014-11-14 2016-05-19 Voyager Therapeutics, Inc. Modulatory polynucleotides
CA3193811A1 (en) 2014-11-14 2016-05-19 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (als)
WO2016094783A1 (en) 2014-12-12 2016-06-16 Voyager Therapeutics, Inc. Compositions and methods for the production of scaav
JP2018506530A (en) 2015-01-30 2018-03-08 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Spinal subpuffy gene delivery system
JP6928558B2 (en) 2015-02-10 2021-09-01 ジェンザイム・コーポレーション Enhanced delivery of viral particles to the striatum and cortex
EP3256588A2 (en) 2015-02-10 2017-12-20 Genzyme Corporation VARIANT RNAi
WO2016131945A1 (en) 2015-02-20 2016-08-25 Transgene Sa Combination product with autophagy modulator
SG11201707116QA (en) 2015-03-10 2017-09-28 Univ Columbia Recombinant glut1 adeno-associated viral vector constructs and related methods for restoring glut1 expression
TWI707951B (en) 2015-04-08 2020-10-21 美商健臻公司 Production of oversized adeno-associated vectors
CA3019315A1 (en) 2015-04-23 2016-10-27 University Of Massachusetts Modulation of aav vector transgene expression
PL3298134T3 (en) 2015-05-16 2023-09-18 Genzyme Corporation Gene editing of deep intronic mutations
SG10202100131WA (en) 2015-06-23 2021-02-25 Childrens Hospital Philadelphia Modified factor ix, and compositions, methods and uses for gene transfer to cells, organs and tissues
MX2018000615A (en) 2015-07-13 2018-08-01 Pivot Bio Inc Methods and compositions for improving plant traits.
BR112018006800A2 (en) 2015-10-05 2018-10-23 Massachusetts Inst Technology nitrogen fixation using restructured nif clusters
WO2017070525A1 (en) 2015-10-22 2017-04-27 University Of Massachusetts Methods and compositions for treating metabolic imbalance in neurodegenerative disease
US20180230489A1 (en) 2015-10-28 2018-08-16 Voyager Therapeutics, Inc. Regulatable expression using adeno-associated virus (aav)
EP3384034B1 (en) 2015-12-02 2020-07-08 The Board of Trustees of the Leland Stanford Junior University Novel recombinant adeno-associated virus capsids with enhanced human skeletal muscle tropism
US10973929B2 (en) 2016-02-03 2021-04-13 The Trustees Of The University Of Pennsylvania Gene therapy for treating mucopolysaccharidosis type I
WO2017139643A1 (en) 2016-02-12 2017-08-17 University Of Massachusetts Anti-angiogenic mirna therapeutics for inhibiting corneal neovascularization
WO2017143100A1 (en) 2016-02-16 2017-08-24 The Board Of Trustees Of The Leland Stanford Junior University Novel recombinant adeno-associated virus capsids resistant to pre-existing human neutralizing antibodies
JP2019512216A (en) 2016-02-22 2019-05-16 ザ・ユニヴァーシティ・オヴ・ノース・キャロライナ・アト・チャペル・ヒル Calcium channel peptide inhibitors
CN115287301A (en) 2016-03-03 2022-11-04 马萨诸塞大学 End-blocked linear duplex DNA for non-viral gene transfer
US11299751B2 (en) 2016-04-29 2022-04-12 Voyager Therapeutics, Inc. Compositions for the treatment of disease
US11326182B2 (en) 2016-04-29 2022-05-10 Voyager Therapeutics, Inc. Compositions for the treatment of disease
WO2017191147A1 (en) 2016-05-04 2017-11-09 Transgene Sa Combination therapy with cpg tlr9 ligand
GB201608046D0 (en) 2016-05-09 2016-06-22 Cambridge Entpr Ltd And Syndey Children S Hospitals Network Randwick And Westmead Incorporating The Treatment of complement-mediated disorders
MX2018014154A (en) 2016-05-18 2019-05-06 Voyager Therapeutics Inc Modulatory polynucleotides.
IL297576B2 (en) 2016-05-18 2024-02-01 Voyager Therapeutics Inc Compositions and methods of treating huntington's disease
CA3027784A1 (en) 2016-06-16 2017-12-21 Charite - Universitatsmedizin Berlin Neuropeptide-expressing vectors and methods for the treatment of epilepsy
MA45493A (en) 2016-06-27 2019-05-01 Aicuris Anti Infective Cures Gmbh HCMC ENTRY INHIBITORS.
CA3029864A1 (en) 2016-07-05 2018-01-11 University Of Massachusetts Aav2-mediated gene delivery of sfasl as a neuroprotective therapy in glaucoma
CA3030156A1 (en) 2016-07-07 2018-01-11 Iovance Biotherapeutics, Inc. Programmed death 1 ligand 1 (pd-l1) binding proteins and methods of use thereof
MX2019000962A (en) 2016-07-26 2019-08-01 Biomarin Pharm Inc Novel adeno-associated virus capsid proteins.
CN110168080A (en) 2016-08-15 2019-08-23 建新公司 The method for detecting AAV
WO2018039375A1 (en) 2016-08-23 2018-03-01 Akouos, Inc. Compositions and methods for treating non-age-associated hearing impairment in a human subject
WO2018049248A1 (en) 2016-09-09 2018-03-15 Icellhealth Consulting Llc Oncolytic virus equipped with bispecific engager molecules
US10457940B2 (en) 2016-09-22 2019-10-29 University Of Massachusetts AAV treatment of Huntington's disease
WO2018060288A1 (en) 2016-09-29 2018-04-05 Glaxosmithkline Biologicals S.A. Compositions and methods of treatment of persistent hpv infection
WO2018069316A2 (en) 2016-10-10 2018-04-19 Transgene Sa Immunotherapeutic product and mdsc modulator combination therapy
TWI788307B (en) 2016-10-31 2023-01-01 美商艾歐凡斯生物治療公司 Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
WO2018091680A1 (en) 2016-11-18 2018-05-24 Transgene Sa Cowpox-based oncolytic vectors
GB201620968D0 (en) 2016-12-09 2017-01-25 Glaxosmithkline Biologicals Sa Adenovirus polynucleotides and polypeptides
KR20190097240A (en) 2016-12-28 2019-08-20 트랜스진 에스.에이. Oncolytic viruses and therapeutic molecules
CN110799474B (en) 2017-01-12 2022-07-26 皮沃特生物公司 Methods and compositions for improving plant traits
CA3059112A1 (en) 2017-04-05 2018-10-11 University Of Massachusetts Cep290 minigene therapy
CA3062450A1 (en) 2017-05-03 2018-11-08 Biomarin Pharmaceutical Inc. Improved lentiviruses for transduction of hematopoietic stem cells
JP2020518258A (en) 2017-05-05 2020-06-25 ボイジャー セラピューティクス インコーポレイテッドVoyager Therapeutics,Inc. Amyotrophic lateral sclerosis (ALS) treatment composition and method
MX2019013172A (en) 2017-05-05 2020-09-07 Voyager Therapeutics Inc Compositions and methods of treating huntington's disease.
US20200123203A1 (en) 2017-06-13 2020-04-23 Flagship Pioneering Innovations V, Inc. Compositions comprising curons and uses thereof
JOP20190269A1 (en) 2017-06-15 2019-11-20 Voyager Therapeutics Inc Aadc polynucleotides for the treatment of parkinson's disease
US11773407B2 (en) 2017-06-26 2023-10-03 Arizona Board Of Regents On Behalf Of Arizona State University CRISPR logic circuits for safer and controllable gene therapies
WO2019010335A1 (en) 2017-07-06 2019-01-10 The Trustees Of The University Of Pennsylvania Aav9-mediated gene therapy for treating mucopolysaccharidosis type i
JP7229989B2 (en) 2017-07-17 2023-02-28 ボイジャー セラピューティクス インコーポレイテッド Trajectory array guide system
WO2019020543A1 (en) 2017-07-28 2019-01-31 Transgene Sa Oncolytic viruses expressing agents targeting metabolic immune modulators
CA3071978A1 (en) 2017-08-03 2019-02-07 Voyager Therapeutics, Inc. Compositions and methods for delivery of aav
CN111356763B (en) 2017-09-22 2024-03-12 建新公司 Variant RNAi
JP2021502060A (en) 2017-10-16 2021-01-28 ボイジャー セラピューティクス インコーポレイテッドVoyager Therapeutics,Inc. Treatment of amyotrophic lateral sclerosis (ALS)
WO2019079242A1 (en) 2017-10-16 2019-04-25 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
JP2021506861A (en) 2017-12-19 2021-02-22 アコーオス インコーポレイテッド AAV-mediated delivery of therapeutic antibodies to the inner ear
EP3762500A1 (en) 2018-03-06 2021-01-13 Voyager Therapeutics, Inc. Insect cell manufactured partial self-complementary aav genomes
US20210023241A1 (en) 2018-03-23 2021-01-28 University Of Massachusetts Gene therapeutics for treating bone disorders
US11608510B2 (en) 2018-03-30 2023-03-21 The Board Of Trustees Of The Leland Stanford Junior University Recombinant adeno-associated virus capsids with enhanced human pancreatic tropism
EP3784290A4 (en) 2018-04-27 2022-03-23 University of Massachusetts Aav capsids identified by in vivo library selection
US20210238630A1 (en) 2018-04-27 2021-08-05 Voyager Therapeutics, Inc. Methods for measuring the potency of aadc viral vectors
CA3098871A1 (en) 2018-05-08 2019-11-14 Rutgers, The State University Of New Jersey Aav-compatible laminin-linker polymerization proteins
PE20210112A1 (en) 2018-05-09 2021-01-19 Biomarin Pharm Inc METHODS OF TREATMENT OF PHENYLKETONURIA
TW202005978A (en) 2018-05-14 2020-02-01 美商拜奧馬林製藥公司 Novel liver targeting adeno-associated viral vectors
WO2019222329A1 (en) 2018-05-15 2019-11-21 Voyager Therapeutics, Inc. Compositions and methods for delivery of aav
KR20210019996A (en) 2018-05-15 2021-02-23 보이저 테라퓨틱스, 인크. Composition and method for the treatment of Parkinson's disease
EP3793615A2 (en) 2018-05-16 2021-03-24 Voyager Therapeutics, Inc. Directed evolution of aav to improve tropism for cns
EP3793686A1 (en) 2018-05-16 2021-03-24 Voyager Therapeutics, Inc. Aav serotypes for brain specific payload delivery
WO2019239311A1 (en) 2018-06-12 2019-12-19 Glaxosmithkline Biologicals Sa Adenovirus polynucleotides and polypeptides
WO2019241486A1 (en) 2018-06-13 2019-12-19 Voyager Therapeutics, Inc. Engineered 5' untranslated regions (5' utr) for aav production
CA3103963A1 (en) 2018-07-02 2020-01-09 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis and disorders associated with the spinal cord
EP3826719A1 (en) 2018-07-24 2021-06-02 Voyager Therapeutics, Inc. Systems and methods for producing gene therapy formulations
TW202022116A (en) 2018-08-03 2020-06-16 美商健臻公司 Variant rnai against alpha-synuclein
US20210380969A1 (en) 2018-10-02 2021-12-09 Voyager Therapeutics, Inc. Redirection of tropism of aav capsids
US20210348242A1 (en) 2018-10-04 2021-11-11 Voyager Therapeutics, Inc. Methods for measuring the titer and potency of viral vector particles
SG11202103425YA (en) 2018-10-05 2021-05-28 Voyager Therapeutics Inc Engineered nucleic acid constructs encoding aav production proteins
US20210371470A1 (en) 2018-10-12 2021-12-02 Voyager Therapeutics, Inc. Compositions and methods for delivery of aav
JP2022505106A (en) 2018-10-15 2022-01-14 ボイジャー セラピューティクス インコーポレイテッド Expression vector for large-scale production of rAAV in the baculovirus / Sf9 system
KR20210082183A (en) 2018-10-25 2021-07-02 리제너론 파아마슈티컬스, 인크. Methods of Analysis of Viral Capsid Protein Compositions
CN113330023A (en) 2018-11-08 2021-08-31 代表亚利桑那州立大学的亚利桑那校董会 Synthetic immune regulation with CRISPR super repressor in vivo
US20220033824A1 (en) 2018-11-29 2022-02-03 University Of Massachusetts Modulation of sptlc1 via recombinant adeno-associated vectors
US11166996B2 (en) 2018-12-12 2021-11-09 Flagship Pioneering Innovations V, Inc. Anellovirus compositions and methods of use
WO2020136232A1 (en) 2018-12-28 2020-07-02 Transgene Sa Immunosuppressive m2 protein
US20220064671A1 (en) 2019-01-18 2022-03-03 Voyager Therapeutics, Inc. Methods and systems for producing aav particles
WO2020172537A1 (en) 2019-02-22 2020-08-27 University Of Massachusetts Oxr1 gene therapy
WO2020190363A1 (en) 2019-03-19 2020-09-24 Massachusetts Institute Of Technology Control of nitrogen fixation in rhizobia that associate with cereals
EP3962536A1 (en) 2019-04-29 2022-03-09 Voyager Therapeutics, Inc. Systems and methods for producing baculoviral infected insect cells (biics) in bioreactors
WO2020227515A1 (en) 2019-05-07 2020-11-12 Voyager Therapeutics, Inc. Compositions and methods for the vectored augmentation of protein destruction, expression and/or regulation
TW202104592A (en) 2019-05-14 2021-02-01 美商拜奧馬林製藥公司 Methods of redosing gene therapy vectors
US20220396806A1 (en) 2019-07-26 2022-12-15 Akouos, Inc. Methods of treating hearing loss using a secreted target protein
EP4010465A1 (en) 2019-08-09 2022-06-15 Voyager Therapeutics, Inc. Cell culture medium for use in producing gene therapy products in bioreactors
TW202122582A (en) 2019-08-26 2021-06-16 美商航海家醫療公司 Controlled expression of viral proteins
WO2021046155A1 (en) 2019-09-03 2021-03-11 Voyager Therapeutics, Inc. Vectorized editing of nucleic acids to correct overt mutations
CN114555814A (en) 2019-09-13 2022-05-27 罗特格斯新泽西州立大学 AAV-compatible laminin-linker polyproteins
JP2022549679A (en) 2019-09-27 2022-11-28 バイオマリン ファーマシューティカル インコーポレイテッド Characterization of Gene Therapy Viral Particles Using Size Exclusion Chromatography and Multi-Angle Light Scattering Techniques
MX2022009252A (en) 2020-01-29 2022-11-09 Genzyme Corp Modified adeno-associated viral capsid proteins for ocular gene therapy and methods of use thereof.
WO2021168362A1 (en) 2020-02-21 2021-08-26 Akouos, Inc. Compositions and methods for treating non-age-associated hearing impairment in a human subject
TW202204377A (en) 2020-03-31 2022-02-01 麻州大學 Capsid variants and uses thereof
US20230131352A1 (en) 2020-04-01 2023-04-27 Voyager Therapeutics, Inc. Redirection of tropism of aav capsids
TW202144579A (en) 2020-04-09 2021-12-01 美商4Mvac有限責任公司 Use of viral vectors for coronavirus vaccine production
CN116249712A (en) 2020-04-15 2023-06-09 沃雅戈治疗公司 TAU binding compounds
GB202005732D0 (en) 2020-04-20 2020-06-03 Synpromics Ltd Regulatory nucleic acid sequences
TW202208406A (en) 2020-05-13 2022-03-01 美商阿科奧斯公司 Compositions and methods for treating kcnq4-associated hearing loss
BR112022023106A2 (en) 2020-05-13 2023-01-17 Voyager Therapeutics Inc AAV CAPSID TROPISM REDIRECT
EP4165195A2 (en) 2020-05-13 2023-04-19 Akouos, Inc. Compositions and methods for treating slc26a4-associated hearing loss
WO2021247995A2 (en) 2020-06-04 2021-12-09 Voyager Therapeutics, Inc. Compositions and methods of treating neuropathic pain
US20230256057A1 (en) 2020-07-13 2023-08-17 Transgene Treatment of immune depression
EP4192514A1 (en) 2020-08-06 2023-06-14 Voyager Therapeutics, Inc. Cell culture medium for use in producing gene therapy products in bioreactors
EP4192515A2 (en) 2020-08-07 2023-06-14 Amicus Therapeutics, Inc. Vesicle targeting proteins and uses of same
CA3190513A1 (en) 2020-09-03 2022-03-10 Jeffrey S. Bartlett Soluble alkaline phosphatase constructs and expression vectors including a polynucleotide encoding for soluble alkaline phosphatase constructs
GB202013940D0 (en) 2020-09-04 2020-10-21 Synpromics Ltd Regulatory nucleic acid sequences
EP4237545A1 (en) 2020-11-02 2023-09-06 BioMarin Pharmaceutical Inc. Process for enriching adeno-associated virus
AU2021391433A1 (en) 2020-12-01 2023-06-22 Akouos, Inc. Anti-vegf antibody constructs and related methods for treating vestibular schwannoma associated symptoms
CA3206361A1 (en) 2020-12-23 2022-06-30 Flagship Pioneering Innovations V, Inc. In vitro assembly of anellovirus capsids enclosing rna
CN117157108A (en) 2020-12-29 2023-12-01 阿库斯股份有限公司 Compositions and methods for treating CLRN1 associated hearing loss and/or vision loss
TW202241479A (en) 2020-12-30 2022-11-01 美商安迅生物製藥公司 Combination therapy of an oncolytic virus delivering a foreign antigen and an engineered immune cell expressing a chimeric receptor targeting the foreign antigen
WO2022187548A1 (en) 2021-03-03 2022-09-09 Voyager Therapeutics, Inc. Controlled expression of viral proteins
EP4301768A2 (en) 2021-03-03 2024-01-10 Voyager Therapeutics, Inc. Controlled expression of viral proteins
TW202309291A (en) 2021-04-07 2023-03-01 法商新植物Sas公司 Compositions and methods for indoor air remediation
CN117242173A (en) 2021-05-03 2023-12-15 安斯泰来再生医药协会 Method for producing mature corneal endothelial cells
TW202321435A (en) 2021-08-11 2023-06-01 美商薩那生物科技公司 Inducible systems for altering gene expression in hypoimmunogenic cells
WO2023034996A1 (en) 2021-09-03 2023-03-09 Biomarin Pharmaceutical Inc. Aav capsid compositions and methods for delivery
WO2023034989A1 (en) 2021-09-03 2023-03-09 Biomarin Pharmaceutical Inc. Aav capsid compositions and methods for delivery
WO2023034997A1 (en) 2021-09-03 2023-03-09 Biomarin Pharmaceutical Inc. Aav capsid compositions and methods for delivery
WO2023034990A1 (en) 2021-09-03 2023-03-09 Biomarin Pharmaceutical Inc. Aav capsid compositions and methods for delivery
WO2023034994A1 (en) 2021-09-03 2023-03-09 Biomarin Pharmaceutical Inc. Aav capsid compositions and methods for delivery
WO2023034980A1 (en) 2021-09-03 2023-03-09 Bomarin Pharmaceutical Inc. Aav capsid compositions and methods for delivery
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
WO2023056329A1 (en) 2021-09-30 2023-04-06 Akouos, Inc. Compositions and methods for treating kcnq4-associated hearing loss
TW202334436A (en) 2021-11-02 2023-09-01 美商航海家醫療公司 Aav capsid variants and uses thereof
TW202334181A (en) 2021-11-17 2023-09-01 美商航海家醫療公司 Aav capsid variants and uses thereof
WO2023107188A1 (en) 2021-12-10 2023-06-15 Massachusetts Institute Of Technology Prediction, biosynthesis, and integration as biosensors of molecules with unique light absorbance signatures and their subsequent in-field remote detection using multi or hyper-spectral cameras
WO2023111580A1 (en) 2021-12-16 2023-06-22 University Of Dundee Targeted degradation of alpha-synuclein
WO2023147374A2 (en) 2022-01-25 2023-08-03 Voyager Therapeutics, Inc. Baculovirus expression system
WO2023150142A1 (en) 2022-02-02 2023-08-10 Akouos, Inc. Anti-vegf antibody constructs and related methods for treating vestibular schwannoma associated symptoms
TW202346599A (en) 2022-02-08 2023-12-01 美商航海家醫療公司 Aav capsid variants and uses thereof
GB202201713D0 (en) 2022-02-10 2022-03-30 Univ Dundee An affinity directed phosphatase system for targeted protein dephosphorylation
WO2023196862A1 (en) 2022-04-06 2023-10-12 Genzyme Corporation Targeted gene therapy for dm-1 myotonic dystrophy
US20230405014A1 (en) 2022-04-12 2023-12-21 Genzyme Corporation Use of an irak4 modulator for gene therapy
WO2023200843A1 (en) 2022-04-12 2023-10-19 The Broad Institute, Inc. Compositions and methods for screening cis regulatory elements
TW202400250A (en) 2022-04-12 2024-01-01 美商健臻公司 Use of irak4 modulators for gene therapy
WO2023201273A1 (en) 2022-04-12 2023-10-19 Genzyme Corporation Dendritic cell assay for innate immunogenicity to gene therapy agents
WO2023213763A1 (en) 2022-05-02 2023-11-09 Transgene Poxvirus encoding a binding agent comprising an anti- pd-l1 sdab
WO2023213764A1 (en) 2022-05-02 2023-11-09 Transgene Fusion polypeptide comprising an anti-pd-l1 sdab and a member of the tnfsf
WO2023220729A2 (en) 2022-05-13 2023-11-16 Flagship Pioneering Innovations Vii, Llc Double stranded dna compositions and related methods
WO2023235791A1 (en) 2022-06-02 2023-12-07 Voyager Therapeutics, Inc. Aav capsid variants and uses thereof
WO2024006741A1 (en) 2022-06-28 2024-01-04 Voyager Therapeutics, Inc. Aav capsid variants and uses thereof
WO2024003353A1 (en) 2022-07-01 2024-01-04 Transgene Fusion protein comprising a surfactant-protein-d and a member of the tnfsf
WO2024011112A1 (en) 2022-07-06 2024-01-11 Voyager Therapeutics, Inc. Aav capsid variants and uses thereof
WO2024009280A1 (en) 2022-07-08 2024-01-11 Baylor College Of Medicine Integrated stress response inhibitors and methods of using the same
WO2024054983A1 (en) 2022-09-08 2024-03-14 Voyager Therapeutics, Inc. Controlled expression of viral proteins
WO2024059739A1 (en) 2022-09-15 2024-03-21 Voyager Therapeutics, Inc. Tau binding compounds
WO2024064856A1 (en) 2022-09-22 2024-03-28 Biomarin Pharmaceutical Inc. Treatment of cardiomyopathy with aav gene therapy vectors
WO2024064863A2 (en) 2022-09-22 2024-03-28 Biomarin Pharmaceutical Inc. Treatment of arrhythmogenic cardiomyopathy with aav gene therapy vectors
WO2024069010A1 (en) 2022-09-30 2024-04-04 Charité - Universitätsmedizin Berlin Gene therapy vectors for the expression of preprodynorphin variants for the treatment of epilepsy
EP4345106A1 (en) 2022-09-30 2024-04-03 Charité - Universitätsmedizin Berlin Gene therapy vectors for the expression of preprodynorphin variants for the treatment of epilepsy

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995013392A1 (en) * 1993-11-09 1995-05-18 Medical College Of Ohio Stable cell lines capable of expressing the adeno-associated virus replication gene

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5135855A (en) * 1986-09-03 1992-08-04 The United States Of America As Represented By The Department Of Health And Human Services Rapid, versatile and simple system for expressing genes in eukaryotic cells
US5478745A (en) * 1992-12-04 1995-12-26 University Of Pittsburgh Recombinant viral vector system
US5591601A (en) * 1993-05-14 1997-01-07 Ohio University Edison Animal Biotechnology Institute DNA polymerase gene expression system utilizing an RNA polymerase co-delivered with the gene expression vector system
DK0733103T3 (en) * 1993-11-09 2004-07-12 Univ Johns Hopkins Preparation of High Titers of Recombinant AAV Vectors
US5693531A (en) * 1993-11-24 1997-12-02 The United States Of America As Represented By The Department Of Health And Human Services Vector systems for the generation of adeno-associated virus particles
EP0785991A1 (en) * 1994-10-13 1997-07-30 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Preparation of rep-negative aav mutants and cells which can be used therefor
US5856152A (en) * 1994-10-28 1999-01-05 The Trustees Of The University Of Pennsylvania Hybrid adenovirus-AAV vector and methods of use therefor
US5872005A (en) * 1994-11-03 1999-02-16 Cell Genesys Inc. Packaging cell lines for adeno-associated viral vectors
EP0796339A1 (en) * 1994-12-06 1997-09-24 Targeted Genetics Corporation Packaging cell lines for generation of high titers of recombinant aav vectors
FR2735789B1 (en) * 1995-06-23 1997-07-25 Centre Nat Rech Scient RECOMBINANT ADENOVIRUSES, THEIR USE FOR PREPARING AVA, COMPLEMENTARY CELL LINE AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995013392A1 (en) * 1993-11-09 1995-05-18 Medical College Of Ohio Stable cell lines capable of expressing the adeno-associated virus replication gene

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LEONARD ET AL. VIROLOGY (1994), 200(2), 566-73 *

Also Published As

Publication number Publication date
CA2264482A1 (en) 1998-03-12
WO1998010088A1 (en) 1998-03-12
EP0931158A1 (en) 1999-07-28
JP2001500015A (en) 2001-01-09
AU4183397A (en) 1998-03-26
IL128780A0 (en) 2000-01-31

Similar Documents

Publication Publication Date Title
AU722624B2 (en) An inducible method for production of recombinant adeno-associated viruses utilizing T7 polymerase
US6274354B1 (en) Methods using cre-lox for production of recombinant adeno-associated viruses
AU723497C (en) Method for recombinant adeno-associated virus-directed gene therapy
WO1998010086A9 (en) Methods using cre-lox for production of recombinant adeno-associated viruses
US20020037867A1 (en) Method for recombinant adeno-associated virus-directed gene therapy
US6251677B1 (en) Hybrid adenovirus-AAV virus and methods of use thereof
AU695811B2 (en) Hybrid adenovirus-AAV virus and methods of use thereof
US20040224411A1 (en) Recombinant adeno-associated virus production
US20040248301A1 (en) Adeno-associated virus vectors with intravector heterologous terminal palindromic sequences
US20100278791A1 (en) Adeno-associated virus serotype i nucleic acid sequences, vectors and host cells containing same
CN1234735A (en) Method for treating skin pigmentation
CN103764831A (en) Capsid-free AAV vectors, compositions, and methods for vector production and gene delivery
CN102002509B (en) Escherichia coli-bacillus subtilis shuttle expression vector and application thereof
WO1992017581A1 (en) Mammalian expression vector
CN101463362B (en) Expression vector for fusion expression of green fluorescent protein, construction method and use thereof
CN109321587B (en) Chimeric antigen receptor T cell interfered by ACAT1 gene
MXPA99002215A (en) Method for recombinant adeno-associated virus-directed gene therapy
CN114032217A (en) Novel coronavirus compound vaccine based on DNA vector and replicative vaccinia virus vector
US20030190746A1 (en) Gene expression control system and its use in recombinant virus packaging cell lines
CA3172664A1 (en) Gene therapy with dysferlin dual vectors
KR20230085912A (en) Recombinant adeno-associated viral vector with CD14 promoter and use thereof
CN116769811A (en) TRAIL-secreting engineered macrophages and uses thereof
CN114921394A (en) Recombinant sheep listeria and use method thereof
MXPA97003105A (en) Hybrid adenovirus-virus aav and mi method of use
CN101343639A (en) Eukaryon expression shuttle vector, construction method and application

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired