AU707866B2 - Packaging cell lines for generation of high titers of recombinant AAV vectors - Google Patents

Packaging cell lines for generation of high titers of recombinant AAV vectors Download PDF

Info

Publication number
AU707866B2
AU707866B2 AU45963/96A AU4596396A AU707866B2 AU 707866 B2 AU707866 B2 AU 707866B2 AU 45963/96 A AU45963/96 A AU 45963/96A AU 4596396 A AU4596396 A AU 4596396A AU 707866 B2 AU707866 B2 AU 707866B2
Authority
AU
Australia
Prior art keywords
aav
cell
packaging
cells
vector
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
AU45963/96A
Other versions
AU4596396A (en
Inventor
James M Allen
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.)
Ampliphi Biosciences Corp
Original Assignee
Targeted Genetics Corp
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 Targeted Genetics Corp filed Critical Targeted Genetics Corp
Publication of AU4596396A publication Critical patent/AU4596396A/en
Application granted granted Critical
Publication of AU707866B2 publication Critical patent/AU707866B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4712Cystic fibrosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/735Fusion polypeptide containing domain for protein-protein interaction containing a domain for self-assembly, e.g. a viral coat protein (includes phage display)
    • 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

Description

WO 96/17947 PCTIJS95/15892 PACKAGING CELL LINES FOR GENERATION
OF
HIGH TITERS OF RECOMBINANT AAV VECTORS Technical Field of the Invention This invention relates to gene therapy, and more specifically to materials and methods used for the generation of high titers of recombinant AAV vectors for use in gene therapy procedures.
Background AAV vectors may have utility for gene therapy but heretofore a significant obstacle has been the inability to generate sufficient quantities of such recombinant vectors in amounts that would be clinically useful for human gene therapy application. This is a particular problem for in vivo applications such as direct delivery to the lung.
Adeno-associated virus (AAV) vectors are among a small number of recombinant virus vector systems which have been shown to have utility as in vivo gene transfer agents (reviewed in Carter, 1992, Current Opinion in Biotechnology, 3:533-539; Muzcyzka, 1992, Curr. Top. Microbiol. Immunol.
158:97-129) and thus are potentially of great importance for human gene therapy. AAV vectors are capable of highfrequency stable DNA integration and expression in a variety of cells including cystic fibrosis (CF) bronchial and nasal epithelial cells (see, Flotte et al., 1992a, Am. J.
Respir. Cell Mol. Biol. 7:349-356; Egan et al., 1992, Nature, 358:581-584; Flotte et al., 1993a, J. Biol. Chem. 268:3781- 3790; and Flotte et al., 1993b, Proc. Natl. Acad. Sci. USA, 93:10163-10167); human bone marrow-derived erythroleukemia cells (see, Walsh et al., 1992, Proc. Natl. Acad. Sci.
WO 96/17947 PCT/US95/15892 2 USA, 89:7257-7261); and several others. AAV may not require active cell division for stable expression which would be a clear advantage over retroviruses, especially in tissue such as the human airway epithelium where most cells are terminally differentiated and non-dividing.
AAV is a defective parvovirus that grows only in cells in which certain functions are provided by a co-infecting helper virus (see Fig. General reviews of AAV may be found in Carter, 1989, Handbook of Parvoviruses, Vol. I, pp. 169-228, Berns, 1990, Virology, pp. 1743-1764, Raven Press, (New York).
Examples of co-infecting viruses that provide helper functions for AAV growth and replication are adenoviruses, herpesviruses and in some cases poxviruses such as vaccinia. The nature of the helper function is not entirely known but appears to be some indirect effect of the helper virus which renders the cell permissive for AAV replication. This belief is supported by the observation that in certain cases AAV replication may occur at a low level of efficiency in the absence of helper virus co-infection if the cells are treated with agents that are either genotoxic or that disrupt the cell cycle.
Although AAV may replicate to a limited extent in the absence of helper virus in certain unusual conditions, as noted above, the more general result is that infection of cells with AAV in the absence of helper functions results in integration of AAV into the host cell genome. The integrated AAV genome may be rescued and replicated to yield a burst of infectious progeny AAV particles if cells containing an integrated AAV provirus are superinfected with a helper virus such as adenovirus. Because the integration of AAV appears to be an efficient event, this suggested that AAV would be a useful vector for introducing genes into cells for stable expression for uses such as human gene therapy.
AAV has a very broad host range with neither any obvious species nor tissue specificity and will replicate in virtually any cell line of human, simian or rodent origin provided an appropriate helper is present. AAV is ubiquitous and has been WO 96/17947 PCTIUS95/15892 3 isolated from a wide variety of animal species including most mammalian and several avian species.
AAV has not been associated with the cause of any disease. AAV is not a transforming or oncogenic virus. AAV integration into chromosomes of human cell lines does not cause any significant alteration in the growth properties or morphological characteristics of the cells. These properties of AAV also recommend it as a potentially useful human gene therapy vector because most of the other viral systems proposed for this application such as retroviruses, adenoviruses, herpesviruses, or poxviruses are disease-causing viruses.
AAV particles are comprised of a protein capsid having three capsid proteins, VPl, VP2, and VP3, and enclosing a DNA genome. The AAV DNA genome is a linear single-stranded
DNA
molecule having a molecular weight of about 1.5 x 106 daltons or approximately 4680 nucleotides long. Strands of either complementary sense, "plus" or "minus" strands, are packaged into individual particles but each particle has only one DNA molecule. Equal numbers of AAV particles contain either a plus or minus strand. Either strand is equally infectious and replication occurs by conversion of the parental infecting single strand to a duplex form and subsequent amplification of a large pool of duplex molecules from which progeny single strands are displaced and packaged into capsids. Duplex or single-strand copies of AAV genomes inserted into bacterial plasmids or phagemids are infectious when transfected into adenovirus-infected cells, and this has allowed the study of AAV genetics and the development of AAV vectors.
The AAV2 genome has two copies of a 145-nucleotide-long ITR (inverted terminal repeat), one on each end of the genome, and a unique sequence region of about 4470 nucleotides long (Srivastava et al., 1983, J. Virol., 45:555-564) that contains two main open reading frames for the rep and cap genes (Hermonat et al., J. Virol. 51:329-339; Tratschin et al., 1984a, J. Virol., 51:611-619). The unique region contains WO 96/17947 PCT/US95/15892 4 three transcription promoters p5, p19, and p40 (Laughlin et al., 1979, Proc. Natl. Acad. Sci. USA, 76:5567-5571) that are used to express the rep and cap genes. The ITR sequences are required in cis and are sufficient to provide a functional origin of replication (ori) and also are sufficient to provide signals required for integration into the cell genome as well as for efficient excision and rescue from host cell chromosomes or from recombinant plasmids. In addition it has been shown that the ITR can function directly as a transcription promoter in an AAV vector (Flotte et al., 1993, vide supra).
The rep and cap genes are required in trans to provide functions for replication and encapsidation of viral genome respectively. The rep gene is expressed from two promoters, p5 and p19. Transcription from p5 yields an unspliced 4.2 kb mRNA which encodes a protein, Rep78, and a spliced 3.9 kb mRNA which encodes a protein, Rep68. Transcription from p19 yields an unspliced mRNA which encodes Rep52 and a spliced 3.3 kb mRNA which encodes Rep40. Thus, the four Rep proteins all comprise a common internal region sequence but differ with respect to their amino and carboxyl terminal regions. Only Rep78 and Rep68 are required for AAV duplex DNA replication, but Rep52 and Rep40 appear to be needed for progeny, singlestrand DNA accumulation. Mutations in Rep78 and Rep68 are phenotypically Rep- whereas mutations affecting only Rep52 and are Rep+ but Ssd-. Rep68 and Rep78 bind specifically to the hairpin conformation of the AAV ITR and possess several enzyme activities required for resolving replication at the AAV termini. Rep52 and Rep40 have none of these properties.
The Rep proteins, primarily Rep78 and Rep68 exhibit several pleiotropic regulatory activities including positive and negative regulation of AAV genes and expression from some heterologous promoters, as well as inhibitory effects on cell growth (Tratschin et al., 1986, Mol. Cell. Biol. 6:2884-2894; Labow et al., 1987, Mol. Cell. Biol., 7:1320-1325; Khleif et al., Virology, 181:738-741). The AAV p5 promoter is WO 96/17947 PCT/US95/15892 5 negatively autoregulated by Rep78 or Rep68 (Tratschin et al., 1986, Mol. Cell. Biol. 6:2884-2894). Because of the inhibitory effects of expression of rep on cell growth, constitutive expression of rep in cell lines has not been readily achieved. For example, Mendelson et al. (1988, Virology, 166:154-165) reported a very low level expression of some Rep proteins in certain cell lines after stable integration of AAV genomes.
The proteins VPl, VP2, and VP3 all share a common overlapping sequence but differ in that VPl and VP2 contain additional amino terminal sequence. All three are coded from the same cap gene reading frame expressed from a spliced 2.3 kb mRNA transcribed from the p40 promoter. VP2 and VP3 are generated from the same mRNA by use of alternate initiation codons. VP1 is coded from a minor mRNA using 3' donor site that is 30 nucleotides upstream from the 3' donor used for the major mRNA that encodes VP2 and VP3. VP1, VP2, and VP3 are all required for capsid production. Mutations which eliminate all three proteins (Cap-) prevent accumulation of singlestrand progeny AAV DNA whereas mutations in the VP1 aminoterminus (Lip-, Inf-) permit single-strand production but prevent assembly of stable infectious particles.
The genetic analysis of AAV that was described-above was based upon mutational analysis of AAV genomes that were molecularly cloned into bacterial plasmids. In early work, molecular clones of infectious genomes of AAV were constructed by insertion of double-strand molecules of AAV into plasmids by procedures such as GC tailing (Samulski et al., 1982, Proc.
Natl. Acad. Sci. USA, 79:2077-2081), addition of synthetic linkers containing restriction endonuclease (Laughlin et al., 1983, Gene, 23:65-73) or by direct, blunt-end ligation (Senapathy Carter, 1984, J. Biol. Chem., 259:4661-4666). It was then shown that transfection of such AAV recombinant plasmids into mammalian cells that were also infected with an appropriate helper virus, such as adenovirus, resulted in rescue and excision of the AAV genome free of any plasmid WO 96/17947 PCT/US95/15892 6 sequence and replication of the rescued genome and generation of a yield of progeny infectious AAV particles. This provided the basis for performing genetic analysis of AAV as summarized above and permitted construction of AAV transducing vectors.
Based on the genetic analysis, the general principles of AAV vector construction were defined as reviewed recently (Carter, 1992, Current Opinions in Biotechnology, 3:533-539; Muzyczka, 1992, Current Topics in Microbiology and Immunology, 158:97-129). AAV vectors are constructed in AAV recombinant plasmids by substituting portions of the AAV coding sequence with foreign DNA to generate a vector plasmid. In the vector plasmid, the terminal (ITR) portions of the AAV sequence must be retained intact because these regions are required in cis for several functions including excision from the plasmid after transfection, replication of the vector genome and integration and rescue from a host cell genome. The vector can then be packaged into an AAV particle to generate an AAV transducing virus by transfection of the vector plasmid into cells that are infected by an appropriate helper virus such as adenovirus or herpesvirus. In order to achieve replication and encapsidation of the vector genome into AAV particles, the vector plasmid must be complemented for any AAV functions required in trans, namely rep and cap, that were deleted in construction of the vector plasmid.
There are at least two desirable features of any AAV vector that is designed for use in human gene therapy. First, the transducing vector must be generated at sufficiently high titers that it is practicable as a delivery system. This is especially important for gene therapy stratagems aimed at in vivo delivery of the vector. It is likely that for many desirable applications of AAV vectors, such as treatment of cystic fibrosis by direct in vivo delivery to the airway, the required dose of transducing vector may be in excess of 1010.
Secondly, the vector preparations must be free of wild-type AAV virus. The attainment of high titers of AAV vectors has been difficult for several reasons including preferential W96/17947 PCT/US95/15892 7 encapsidation of wild-type AAV genomes if they are present or generated by recombination, and the inability to generate sufficient complementing functions such as rep or cap. Useful cell lines expressing such complementing functions have not been generated, in part, because of several inhibitory functions of the rep gene.
The first AAV vectors that were described contained foreign reporter genes such as neo or cat or dhfr that were expressed from AAV transcription promoters or an SV40 promoter (Tratschin et al., 1984b, Mol. Cell. Biol. 4:2072-2081; Hermonat Muzyczka, 1984, Proc. Natl. Acad. Sci. USA, 81:6466-6470; Tratschin et al., 1985, Mol. Cell. Biol. 5:3251- 3260; McLaughlin et al., 1988, J. Virol., 62:1963-1973; Lebkowski et al., 1988 Mol. Cell. Biol., 7:349-356). These vectors were packaged into AAV-transducing particles by cotransfection into adenovirus-infected cells together with a second packaging plasmid that contained the AAV rep and cap genes expressed from the natural wild-type AAV transcription promoters. In an attempt to prevent packaging of the packaging plasmid into AAV particles several approaches were taken. In some cases, (Hermonat Muzyczka, 1984; McLaughlin et al., 1988) the packaging plasmid had inserted a large region of bacteriophage lambda DNA within the AAV sequence to generate an oversized genome that could not be packaged. In other cases, (Tratschin et al., 1984b; Tratschin et al., 1985, Lebkowski et al., 1988), the packaging plasmid had deleted the ITR regions of AAV in order that it could not be excised and replicated and thus could not be packaged. All of these approaches failed to prevent generation of particles containing wild-type AAV DNA and also failed to generate effective high titers of AAV transducing particles. Indeed titers of not more than 104 ml were cited by Hermonat Muzyczka, 1984. The production of wild-type AAV particles in these studies was probably due to the presence of overlapping homology between AAV sequences present in the vector and packaging plasmids. It was shown by Senapathy and Carter WO 96/17947 PCT US95/15892 8 (1984, J. Biol. Chem. 259:4661-4666) that the degree of recombination in such a system is approximately equivalent to the degree of sequence overlap. It was suggested in a review of the early work (Carter 1989, Handbook of Parvoviruses, Vol.
II, pp. 247-284, CRC Press, Boca Raton, FL) that titers of 106 per ml might be obtained, but this was based on the abovecited studies in which large amounts of wild-type AAV contaminated the vector preparation. Such vector preparations containing wild-type AAV are not useful human gene therapy.
Furthermore, these early vectors exhibited low transduction efficiencies and did not transduce more than 1 or 2% of cells in cultures of various human cell lines even though the vectors were supplied at multiplicities of up to 50,000 particles per cell. This may have reflected in part the contamination with wild-type AAV particles and the presence of the AAV rep gene in the vector. Furthermore, Samulski et al.
(1989, J. Virol. 63:3822-3828) showed that the presence of wild-type AAV significantly enhanced the yield of packaged vector. Thus, in packaging systems where the production of wild-type AAV is eliminated, the yield of packaged vector may actually be decreased. Nevertheless, for use in any human clinical application it will be essential to eliminate production of wild-type AAV.
Additional studies (McLaughlin et al., 1988; Lebkowski et al., 1988) to generate AAV vectors which did not contain the AAV rep or cap gene still met with generation of wild-type AAV and still produced very low transduction frequencies on human cell lines. Thus, McLaughlin et al., 1988 reported that AAV rep- cap- vectors containing the neo gene packaged with the same packaging plasmid used earlier by Hermonat Muzyczka (1984) still contained wild-type AAV. As a consequence it was only possible to use this virus at a multiplicity of 0.03 Sparticles per cell 300 infectious units per 10,000 cell) to avoid double hits with vector and wild-type particles. When the experiment was done in this way, by infecting 32,000 cells with 1000 infectious units, an average WO 96/17947 PCT/US95/15892 9 of 800 geneticin-resistant colonies was obtained. Although this was interpreted as demonstrating the virus was capable of yielding a transduction frequency of 80%, in fact only 2.5% of the cells were transduced. Thus the effectively useful titer of this vector was limited. Furthermore, this study did not demonstrate that the actual titer of the vector preparation was any higher than those obtained previously by Hermonat Muzyczka (1984). Similarly, Lebkowski et al., 1988, packaged AAV vectors which did not contain either a rep or cap gene and used an ori- packaging plasmid pBalA identical to that used earlier by Tratschin et al., (1984b, 1985) and reported transduction frequencies that were similarly low, in that for several human cell lines not more than 1% of the cells could be transduced to geneticin resistance even with their most concentrated vector stocks. Lebkowski et al., (1988) did not report the actual vector titers in a meaningful way but the biological assays showing not more than 1% transduction frequency when 5 x 106 cells were exposed to three ml of vector preparation indicates that the titer was less than 2 x 10 4 Also, the pBal packaging plasmid contains overlapping homology with the ITR sequence in the vector and leads to generation by recombination of wild-type AAV.
Laface et al., (1988) used the same vector as that used by Hermonat Muzyczka (1984) prepared in the same way and obtained a transduction frequency of 1.5% in murine bone marrow cultures again showing very low titer.
Samulski et al., (1987, J. Virol., 61:3096-3101) constructed a plasmid called pSub201 which was an intact AAV genome in a bacterial plasmid but which had a deletion of 13 nucleotides at the extremity of each ITR and thus was rescued and replicated less efficiently than other AAV plasmids that contained the entire AAV genome. Samulski et al. (1989, J.
SVirol., 63:3822-3828) constructed AAV vectors based on pSub201 but deleted for rep and cap and containing either a hyg or neo gene expressed from an SV40 early gene promoter. They packaged these vectors by co-transfection with a packaging WO 96/17947 917947 PCT/US95/15892 10 plasmid called pAAV/Ad which consisted of the entire AAV nucleotide sequence from nucleotide 190 to 4490 enclosed at either end with one copy of the adenovirus ITR. In this packaging plasmid the AAV rep and cap genes were expressed from the natural AAV promoters p5, p19 and p40. The function of the adenovirus ITR in pAAV/Ad was thought to be to enhance the expression level of AAV capsid proteins. However, rep is expressed from its homologous promoter and is negatively regulated and thus its expression is limited. Using their encapsidation system Samulski et al., 1989, generated AAV vector stocks that were substantially free of wild-type AAV but had transducing titers of only 3 x 10 4 hygromycin-resistant units per ml of supernatant. When a wild-type AAV genome was used in the packaging plasmid the titer of the AAV vector prep was increased to 5 x 104. The low titer produced in this system thus appears to have been due in part to the defect in the ITR sequences of the basic pSub201 plasmid used for vector construction and in part due to limiting expression of AAV genes from pAAV/Ad. In an attempt to increase the titer of the AAVneo vector preparation, Samulski et al., 1989, generated vector stocks by transfecting, in bulk, thirty dishes of 293 cells and concentrating the vector stock by banding in CsCl. This produced an AAVneo vector stock containing a total of 108 particles as measured by a DNA dotblot hybridization assay. When this vector stock was used at multiplicities of up to 1,000 particles per cell, a transduction frequency of 70% was obtained. This suggests that the particle-to-transducing ratio is about 500 to 1,000 particles since at the ratio of one transducing unit per cell the expected proportion of cells that should be transduced is 63% according to the Poisson distribution.
Although the system of Samulski et al., 1989, using the vector plasmid pSub201 and the packaging plasmid pAAV/Ad did not have overlapping AAV sequence homology between the two plasmids, there is overlapping homology at the XbaI sites and recombination of these sites leads to generation of complete WO 96/17947 PCT/US95/15892 11 wild-type AAV. That is, although overlapping homology of AAV sequence is not present, the complete AAV sequence is contained within the two plasmids, and thus recombination can generate wild-type AAV, which is undesirable. That this class of recombination occurs in AAV plasmids was shown by Senapathy Carter (1984, J. Biol. Chem. 259:4661-4666). Therefore, because of the problems of low titer and ability to generate wild-type recombinants, the system described by Samulski et al., 1989, does not have utility for human gene therapy.
Several other reports have described AAV vectors.
Srivastava et al., (1989, Proc. Natl. Acad. Sci. USA, 86:8078-8082) described an AAV vector based on the pSub201 plasmid of Samulski et al., (1987), in which the coding sequences of AAV were replaced with the coding sequences of another parvovirus, B19. This vector was packaged into AAV particles using the pAAV/Ad packaging plasmid and generated a functional vector, but titers were not reported. This system was based on pSub201 and thus suffers from the defect described above for this plasmid. Second, the vector and the packaging plasmid both contained overlapping AAV sequences (the ITR regions) and thus recombination to give contaminating wild-type virus is highly likely.
Chatterjee et al. (1991, Vaccines 91, Cold Spring Harbor Laboratory Press, pp. 85-89), Wong et al. (1991 Vaccines 91, Cold Spring Harbor Laboratory Press, pp. 183-189), and Chatterjee et al. (1992, Science, 258:1485-1488) describe AAV vectors designed to express antisense RNA directed against infectious viruses such as HIV or Herpes simplex virus.
However, these authors did not report any titers of their AAV vector stocks. Furthermore, they packaged their vectors using an Ori- packaging plasmid analogous to that used by Tratschin et al. (1984b, 1985) containing the BalA fragment of the AAV genome and therefore their packaging plasmid contained AAV vector sequences that have homology with AAV sequences that were present in their vector constructs. This will also lead to generation of wild-type AAV. Thus, Chatterjee et al., and WO9617947 PCTIUS95/15892 12 Wong et al., used a packaging system known to give only low titer and which can lead to generation of wild-type AAV genomes because of the overlapping homology in the vector and packaging sequences.
Other reports have described the use of AAV vectors to express genes in human lymphocytes (Muro-Cacho et al., 1992, J. Immunotherapy, 11:231-237) or a human erythroid leukemia cell line (Walsh et al., 1992, Proc. Natl. Acad. Sci. USA, 89:7257-7261) with vectors based on the pSub201 vector plasmid and pAAV/Ad packaging plasmid. Again, titers of vector stocks were not reported and were apparently low because a selective marker gene was used to identify those cells that had been successfully transduced with the vector.
Transduction of human airway epithelial cells, grown in vitro from a cystic fibrosis patient, with an AAV vector expressing the selective marker gene neo from the AAV promoter was reported (Flotte et al., 1992, Am. J. Respir.
Cell. Mol. Biol. 7:349-356). In this study the AAVneo vector was packaged into AAV particles using the pAAV/Ad packaging plasmid. Up to 70% of the cells in the culture could be transduced to geneticin resistance and the particle-totransducing ratio was similar to that reported by Samulski et al., (1989). Thus to obtain transduction of 70% of'the cells, a multiplicity of up to several hundred vector particles per cell was required. Transduction of human airway epithelial cells in in vitro culture using an AAV transducing vector that expressed the CFTR gene from the AAV ITR promoter showed that the cells could be functionally corrected for the electrophysiological defect in chloride channel function that exists in cells from cystic fibrosis patients (Egan et al., Nature, 1992, 358:581-584; Flotte et al., J. Biol. Chem.
268:3781-3790).
The above-cited studies suggest that AAV vectors may have potential utility as vectors for treatment of human disease by gene therapy. However, the ability to generate sufficient amounts of AAV vectors has been a severe limitation on the WO 96/17947 PCTIUS95/15892 13 development of human gene therapy using AAV vectors. One aspect of this limitation is that there have been very few studies using AAV vectors in in vivo animal models (see, e.g., Flotte et al., 1993b; and Kaplitt et al., 1994, Nature Genetics 8:148-154). This is generally a reflection of the difficulty associated with generating sufficient amounts of AAV vector stocks having a high enough titer to be useful in analyzing in vivo delivery and gene expression. One of the limiting factors for AAV gene therapy has been the relative inefficiency of the vector packaging systems that have been used. Because of the lack of cell lines expressing the AAV trans complementing functions, such as rep and cap, packaging of AAV vectors has been achieved in adenovirus-infected cells by co-transfection of a packaging plasmid and a vector plasmid. The efficiency of this process may be limited by the efficiency of transfection of each of the plasmid constructs, and by the level of expression of Rep proteins from the packaging plasmids described to date. Each of these problems appears to relate to the biological activities of the AAV Rep proteins. In addition, as noted above, all of the packaging systems described above have the ability to generate wild-type AAV by recombination.
The lack of cell lines stably expressing functional Rep apparently reflects a cytotoxic or cytostatic function of Rep as shown by the inhibition by Rep of neo-resistant colony formation (Labow et al., 1987; Trempe et al., 1991). This also appears to relate to the tendency of Rep to reverse the immortalized phenotype in cultured cells, which has made the production of cell lines stably expressing functional Rep extremely difficult. Several attempts to generate cell lines expressing Rep have been made. Mendelson et al., (1988, Viroloqy, 166:154-165) reported obtaining in one cell line some low level expression of AAV Rep52 protein but no Rep78 or Rep68 protein after stable transfection of HeLa or 293 cells with plasmids containing an AAV rep gene. Because of the absence of Rep78 and Rep68 proteins, vector could not be WO 96/17947 9617947 PCT/US95/15892 14 produced in the cell line. Another cell line made a barely detectable amount of Rep78 which was nonfunctional.
Vincent et al. (1990, Vaccines 90, Cold Spring Harbor Laboratory Press, pp. 353-359) attempted to generate cell lines containing the AAV rep and cap genes expressed from the normal AAV promoters, but these attempts were not successful either because the vectors were contaminated with a 100-fold excess of wild-type AAV particles or because the vectors were produced at only very low titers of less than 4 x 10 3 In an alternate approach, Lebkowski et al. patent 5,173,414, issued Dec. 22, 1992) constructed cell lines containing AAV vectors in an episomal plasmid. These cell lines could then be infected with adenovirus and transfected with the trans complementing AAV functions rep and cap to generate preparations of AAV vector. It is claimed that this allows higher titers of AAV stocks to be produced. However, in the examples shown, the only information relative to titer that is shown is that one human cell line, K562, could be transduced at efficiencies of only 1% or less, which does not indicate high titer production of any AAV vector. In this system the vector is carried as an episomal (unintegrated construct), and it is stated that integrated copies of the vector are not preferred. In a subsequent patent No.
5,354,678, issued October 11, 1994), Lebkowski et al.
introduce rep and cap genes into the cell genome but the method again requires the use of episomal AAV transducing vectors comprising an Epstein-Barr virus nuclear antigen (EBNA) gene and an Epstein-Barr virus latent origin of replication; and, again, the only information relative to titer showed a fairly low titer.
The approach to packaging of AAV vectors described by Lebkowski et al., 1992, has several undesirable aspects.
First, maintaining the vector as an unintegrated, high copy number episomal plasmid in a cell line is not desirable because the copy number per cell cannot be rigorously controlled and episomal DNA is much more likely to undergo WO 96/17947 PCT'US95/15892 15 rearrangement leading to production of defective vectors.
Secondly, in this system, the vector must still be packaged by infecting the cell line with adenovirus and introducing a plasmid containing the AAV rep and cap genes. The plasmid used by Lebkowski et al., 1992, again was pBal which, as noted above, has overlapping homology with the vector ITR sequences and will result in generation of wild-type AAV. Third, in the pBal packaging plasmid used by Lebkowski et al., 1988, 1992, the rep gene is expressed off its homologous p5 promoter and is thus negatively autoregulated and therefore rep expression is likely to be limited.
The problem of suboptimal levels of rep expression after plasmid transfection may relate to another biological activity of these proteins. There is evidence (Tratschin et al., 1986, Mol. Cell. Biol. 6:2884-2894) that AAV-Rep proteins downregulate their own expression from the AAV-p5 promoter which has been used in all of the previously described packaging constructs such as pAAV/Ad (Samulski et al., 1989) or pBal (Lebkowski et al., 1988, 1992).
Another attempt to develop cell lines expressing functional rep activity was recently published by H61scher et al. (1994, J. Virol. 68:7169-7177). They described the generation of cell lines in which rep was placed under control of a glucocorticoid-responsive MMTV promoter. Although they observed particle formation, the particles were apparently noninfectious. Additional experiments indicated that the defect was quite fundamental; namely, there was virtually no accumulation of single-stranded rAAV DNA in the cells.
Production of infectious particles required an additional transient transfection with constitutive highly-expressed rep constructs they had to "add back" rep activity to cells that were supposed to be able to provide it themselves).
Summary of the Invention One of the basic challenges for gene therapy has been the development of strategies for transduction of cells and tissues which cannot be easily manipulated ex vivo or which are not actively dividing. AAV vectors can achieve in vivo gene transfer in the respiratory tract, for example, but high titers are critical so as to allow for the delivery of sufficiently high multiplicity of vector in as small a volume as possible. This makes optimal packaging methodology of central importance in determining the feasibility of an AAV-based gene therapy. Stable, helper-free AAV packaging cell lines have been elusive, mainly due to the activities of Rep protein, which downregulates its own expression and can negatively affect the host cell. The approaches described in this invention effectively circumvent these problems and have allowed for substantial improvements in packaging efficiency.
A number of preferred embodiments of the present invention are summarized below: 1. A method of producing a mammalian cell for packaging of a recombinant AAV (rAAV) vector, said method including the steps of: providing a mammalian cell which includes a stably integrated AAV cap gene operably linked to a promoter, and a stably integrated AAV rep gene operably linked to a helper-virus inducible heterologous 2 promoter, wherein p5 promoter function has been replaced by the helper-virus inducible heterologous promoter; .5 replicating the cell of step to produce a population of cells; and introducing a helper virus to the population of cells of step wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep gene.
2. A method according to claim 1, wherein said helper virus is an adenovirus.
3. A method according to claim 1, wherein said packaging cell grows at least one half as rapidly as parental-type cells that do not contain an AAV rep gene, and wherein said packaging cell when used to package rAAV vectors produces at least 100 rAAV particles/cell.
4. A method according to claim 1, wherein said mammalian cell of step includes the combined rep and cap genes of AAV in which the promoter has been replaced by a heterologous promoter.
A method according to claim 4, wherein said heterologous promoter is a mouse metallothionein I (mMT-I) promoter.
6. A cell produced by the method of claim 1, and progeny thereof, wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep gene.
7. A cell produced by the method of claim 3, and progeny thereof, wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep gene.
8. A cell produced by the method of claim 4, and progeny thereof, 20 wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep gene.
9. A cell produced by the method of claim 5, and progeny thereof, wherein said cell exhibits helper virus-inducible expression of said stably 25 integrated AAV rep gene.
A mammalian cell for packaging of a recombinant AAV (rAAV) vector, :"*said cell including a stably integrated cap gene operably linked to a promoter, and a stably integrated rep gene operably linked to a helper virus-inducible heterologous promoter; wherein p5 promoter function has been replaced by the helper virus-inducible heterologous promoter and wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep gene.
11. An AAV packaging cell of claim 10, wherein said helper-virusinducible expression of said stably integrated AAV rep gene is inducible by adenovirus.
12. An AAV packaging cell of claim 10, wherein said packaging cell grows at least one half as rapidly as parental-type cells that do not contain an AAV rep gene, and wherein said packaging cell when used to package rAAV vectors produces at least 100 rAAV particles/cell.
13. An AAV packaging cell of claim 10, wherein said cell includes the combined rep and cap genes of AAV in which the p5 promoter has been replaced by a heterologous promoter.
14. An AAV packaging cell of claim 13, wherein said heterologous promoter is a mouse metallothionein I (mMT-I) promoter.
15. An AAV packaging cell of claim 10, further including a stably integrated recombinant AAV (rAAV) vector, said vector including a 20 polynucleotide sequence of interest located between two AAV inverted terminal repeat (ITR) regions, wherein said polynucleotide is operably linked to a promoter.
s* 16. A method of packaging a recombinant AAV vector, including the steps 25 of: providing an AAV packaging cell of claim introducing a recombinant AAV (rAAV) vector, said vector including a polynucleotide sequence of interest located between two AAV inverted terminal repeat (ITR) regions, wherein said polynucleotide is operably linked to a promoter; introducing a helper virus; and incubating the cell under conditions suitable for replication and packaging of AAV such that said rAAV vector is packaged.
18A 17. A method of packaging a recombinant AAV vector, including the steps of: providing an AAV packaging cell of claim 15 which includes a stably integrated rAAV vector including a polynucleotide of interest operably linked to a promoter; introducing a helper virus; and incubating the cell under conditions suitable for replication and packaging of AAV such that said rAAV vector is packaged.
18. A method of determining the relative infectious titer of an rAAV vector preparation, including the steps of: introducing a helper virus and serial dilutions of the rAAV vector preparation to AAV packaging cells of claim 0 incubating the cells under conditions suitable for replication of AAV; 20 and determining the amount of replicated rAAV vector relative to an rAAV preparation of known titer.
25 19. The method of claim 1, further including the step of selecting a cell S.. exhibiting helper-virus-inducible expression of said stably integrated AAV rep gene.
Throughout this specification the word "comprise", or variations such 30 as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
WO 96/17947 917947 PCTIUS95/15892 19 Brief Description of the Drawings Figure 1 is a diagram of plasmid pMt-rep/cap//pKO-neo, as described in Example 1.
Figure 2 is a reproduction of Southern blots demonstrating that packaging cells produced according to the present invention have sufficient rep activity to replicate an incoming rAAV vector, as described in Example 4.
Figure 3 is a reproduction of Southern blots demonstrating that packaging cells produced according to the present invention are capable of replicating an AAV genome in the presence of adenovirus, and that this activity can be used to quantify the number of infectious viral particles present in a given sample, as described in Example Figure 4 is a reproduction of Southern blots demonstrating that packaging cells produced according to the present invention express rep protein and are able to replicate recombinant AAV plasmid DNA genomes introduced by transfection, as described in Example 6.
Figure 5 is a reproduction of Southern blots demonstrating that the infectious rAAV titering assay described in Example 5 had proceeded for a sufficient amount of time to reach a maximum, as described in Example'7.
Figures 6 and 7 are reproductions of Southern blots demonstrating that packaging cells produced according to the present invention can replicate and package rAAV vector genomes into infectious virions by either transfection or infection, as described in Examples 9 and Figure 8 is a reproduction of a Southern blot demonstrating that packaging cells produced according to the present invention possess sufficient rep activity to recognize, excise and amplify an integrated rAAV vector, as described in Example 11.
WO 96/17947 PCT/US95/15892 20 DETAILED DESCRIPTION OF THE INVENTION AAV vectors are recombinant constructs of the AAV virus comprising AAV components necessary for replication and encapsidation, along with a heterologous polynucleotide encoding a protein of interest. These recombinant AAV vectors are potentially powerful tools for human gene therapy, particularly for diseases such as cystic fibrosis and sickle cell anemia. A major advantage of AAV vectors over other approaches to gene therapy is that they do not require ongoing replication of the target cell in order to integrate permanently into the cell's genome.
The invention described herein provides methods and materials for use in the production of high titers of recombinant AAV vectors for use in gene therapy. It also provides methods and materials for determining the relative infectious titer of rAAV preparations.
The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See Sambrook, Fritsch, and Maniatis, Molecular Cloning: A Laboratory Manual, Second Edition (1989), Oliconucleotide Synthesis Gait Ed., 1984), Animal Cell Culture Freshney, Ed., 1987), the series Methods in Enzvmoloqy (Academic Press, Inc.); Gene Transfer Vectors for Mammalian Cells Miller and M.P.
Calos eds. 1987), Handbook of Experimental Immunology,
(D.M.
Weir and C.C. Blackwell, Eds.), Current Protocols in Molecular Biology Ausubel, R. Brent, R.E. Kingston, D.D. Moore, J.G. Siedman, J.A. Smith, and K. Struhl, eds., 1987), and Current Protocols in Immunology Coligan, A.M. Kruisbeek, D.H. Margulies, E.M. Shevach and W. Strober, eds., 1991). All patents, patent applications, and publications mentioned herein, both supra and infra, are hereby incorporated herein by reference.
WO 96/17947 PCT/US95/15892 21 Definitions: The terms "polypeptide", "peptide" and "protein" are used interchangeably to refer to polymers of amino acids of any length. These terms also include proteins that are posttranslationally modified through reactions that include glycosylation, acetylation and phosphorylation.
"Polynucleotide" refers to a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides, or analogs thereof. This term refers only to the primary structure of the molecule. Thus, doubleand single-stranded DNA, as well as double- and singlestranded RNA are included. It also includes modified polynucleotides such as methylated or capped polynucleotides.
"Recombinant," as applied to a polynucleotide, means that the polynucleotide is the product of various combinations of cloning, restriction and/or ligation steps, and other procedures that result in a construct that is distinct from a polynucleotide found in nature.
"Sequence overlap" between two polynucleotides occurs when the nucleotides share a homologous sequence of sufficient length and identity that recombination is facilitated. The level of homology and corresponding frequency of recombination increase with increasing length of the homologous sequences and with their level of shared identity. The level of homology that will pose a concern in a given system can be determined theoretically and confirmed experimentally, as is known in the art. Typically, however, recombination can be substantially reduced or eliminated if the overlapping sequence is less than about a 25 nucleotide sequence if it is at least 80% identical over its entire length, or less than about a 50 nucleotide sequence if it is at least 70% identical over its entire length. Of course, even lower levels of homology are preferable since they will further reduce the likelihood of recombination.
A "vector" refers to a recombinant plasmid or virus that comprises a polynucleotide to be delivered into a host cell, WO 96/17947 PCT/US95/15892 22 either in vitro or in vivo. The polynucleotide to be delivered, sometimes referred to as a "target polynucleotide", may comprise a coding sequence of interest in gene therapy.
A "recombinant AAV vector" (or "rAAV vector") refers to a vector comprising one or more polynucleotides of interest that are flanked by AAV inverted terminal repeat sequences (ITRs).
Such rAAV vectors can be replicated and packaged into infectious viral particles when present in a host cell that has been infected with a suitable helper virus and is expressing the AAV rep and cap genes.
AAV "rep" and "cap" genes (encoding replication and encapsidation proteins, respectively) have been found in all AAV serotypes examined, and are described above and in the references cited therein. Typically, the rep and cap genes are found adjacent to each other in the AAV genome, and they are generally conserved among AAV serotypes.
A "helper virus" for AAV refers to a second virus that allows wild-type AAV (which is a "defective" parvovirus) to be replicated and packaged by a host cell. A number of such helper viruses have been identified, including adenoviruses, herpesviruses and poxviruses such as vaccinia.
"Packaging" as used herein refers to a series of subcellular events that results in the assembly and' encapsidation of an rAAV vector. Thus, when a suitable vector plasmid is introduced into a packaging cell line under appropriate conditions, it will be assembled into a vector viral particle.
"Heterologous" means derived from a genotypically distinct entity from that of the rest of the entity to which it is compared. For example, a polynucleotide introduced by genetic engineering techniques into a different cell type is a heterologous polynucleotide (and, when expressed, can encode a heterologous polypeptide). Similarly, a promoter that is removed from its native coding sequence and operably linked to a different coding sequence is a heterologous promoter.
WO 96/17947 PCTUS95/15892 23 "Promoter", as used herein, refers to a genomic region that enhances the transcription of a gene or coding sequence to which it is operably linked.
"Operably linked" refers to a juxtaposition, wherein the components so described are in a relationship permitting them to function in their intended manner. A promoter is operably linked to a coding sequence if the promoter promotes transcription of the coding sequence. An operably linked promoter is usually in cis configuration with the coding sequence, but is not necessarily contiguous with it.
"Host cells", "cell lines", "cell cultures", and other such terms denote higher eukaryotic cells, most preferably mammalian cells, which can be used as recipients for recombinant vectors or other transfer polynucleotides, and include the progeny of the original cell that was transduced.
It is understood that the progeny of a single cell may not necessarily be completely identical (in morphology or in genomic complement) to the original parent cell.
"Stable integration" of a polynucleotide into a cell means that the polynucleotide has been introduced into a chromosome or mini-chromosome of the cell and, therefore, becomes a relatively permanent part of the cellular genome.
Although "episomes" such as plasmids can sometimes be maintained for many generations (particularly if kept under selective pressure), genetic material carried episomally is generally more susceptible to loss than chromosomallyintegrated material. Also, the chromatin structure of eukaryotic chromosomes can influence the level of expression of an integrated polynucleotide; and we believe that such effects can sometimes prove beneficial in situations such as those described herein (in which the level of expression of the AAV rep gene can have negative effects upon cellular metabolism). The selection of stable cell lines having properties that are particularly desirable in the context of the present invention, are described in the Detailed Description and Examples below.
WO 96/17947 PCT/US95/15892 24 "Efficiency" when used in describing a cell line refers to the useful properties of the line; in particular, the growth rate, and (for packaging cell lines) the number of virus particles produced per cell. "High efficiency packaging" indicates production of at least 100 viral particles per cell.
Modes of carrying out the invention: The method for producing high titers of recombinant.AAV vectors comprises several steps. The general strategy involves preparation of mammalian packaging cell lines that comprise a stably integrated AAV cap gene operably linked to a promoter, and a stably integrated AAV rep gene operably linked to a heterologous promoter. Packaging cells are then infected or transfected with a plasmid comprising the AAV ITR regions and the target polynucleotide. Under suitable conditions (including suitable growth conditions and infection with a competent helper virus), expression of the rep and cap genes of the packaging cell results in the synthesis of rep and cap proteins which mediate replication and encapsidation of the AAV vector, respectively. Providing a polynucleotide of interest (also referred to as a "target polynucleotide") inbetween the AAV ITR sequences of the rAAV vector, thus results in packaging of the target polynucleotide into an infectious rAAV particle which can be used to deliver the polynucleotide to a desired host cell.
By minimizing the extent of sequence overlap between the AAV genes in the packaging cell line and those of the vector plasmid, the proportion of wild-type AAV particles not containing the target polynucleotide) can be minimized. As described in the Background section, the presence of contaminating wild-type AAV limits the therapeutic potential of rAAV vector preparations. Besides the lack of sequence overlap in the constructs of the present invention, the promoter region is replaced with a different promoter. The packaging cell lines of the present invention enable the WO 96/17947 PCT/US95/15892 25 efficient production of rAAV preparations that are of high titer and are substantially free of any contaminating wildtype AAV; attributes that are especially useful in the context of AAV-mediated gene therapy.
Although a first illustration of the principles of the present invention was performed using the AAV2 serotype, it is expected that these same principles will be applicable to other AAV serotypes since it is now known that the various serotypes are quite closely related both functionally and structurally, even at the genetic level (see, Blacklow, 1988, pp. 165-174 of "Parvoviruses and Human Disease",
J.R.
Pattison and Rose, 1974, Comprehensive Virology 3: 1-61). For example, all AAV serotypes apparently exhibit very similar replication properties mediated by homologous rep genes; and all bear three related capsid proteins such as those expressed in AAV2. The degree of relatedness is further suggested by heteroduplex analysis which reveals extensive cross-hybridization between serotypes along the length of the genome; and the presence of analogous self-annealing segments at the termini that correspond to ITRs. The similar infectivity patterns also suggest that the replication functions in each serotype are under similar regulatory control.
Producing the packaging cell line: The parental lines from which packaging cells are generated may be obtained from any cell line that is susceptible to AAV infection, and amenable to culture in vitro. As indicated earlier, AAV has a very broad host range and has been isolated from a variety of mammalian cell types, including simian, human and rodent cells. For human gene therapy, human cell lines in which appropriate helper functions can be expressed are typically preferred. Such human cell lines from which the packaging cell lines may be derived, include, for example, Hela, A549, 293, KB, Detroit, and WI38 cells. We initially selected both Hela cells and WO 96/17947 PCTIUS95/15892 26 A549 cells for demonstrations of the present invention. As described in the Examples below, we were readily able to generate packaging cells from both parental lines tested.
In the case of wild-type AAV (using AAV2 for purposes of illustration), the rep gene is under regulation of the promoter, which is itself strongly down-regulated by rep expression. In constructing packaging cell lines according to the present invention, the cells are provided with a stably integrated AAV cap gene operably linked to a promoter, and a stably integrated AAV rep gene operably linked to a heterologous promoter; as described and illustrated herein.
Any heterologous promoter that is not strongly down-regulated by rep gene expression is suitable; but inducible promoters are preferred because constitutive expression of the rep gene can have a negative impact on the host cell. Since the rep and cap genes of the present invention are to be stably integrated into the host cell genome, we believe that location effects (possibly due to chromatin structure) can also influence expression of the genes and can be taken advantage of in obtaining preferred packaging lines. In particular, the methodology described below can be used to generate and select packaging cells that exhibit the desired properties. A large variety of inducible promoters are known in the art; including, by way of illustration, heavy metal ion inducible promoters (such as metallothionein promoters); steroid hormone inducible promoters (such as the MMTV promoter or growth hormone promoters); and promoters such as those from T7 phage which are active in the presence of T7 RNA polymerase.
An especially preferred sub-class of inducible promoters are those that are induced by the helper virus that is used to complement the replication and packaging of the rAAV vector.
Anumber of helper-virus-inducible promoters are known, including for example, the adenovirus early gene promoter which is inducible by adenovirus EIA protein; the adenovirus major late promoter; the herpesvirus promoter which is WO 96/17947 PCT/US95/15892 27 inducible by herpesvirus proteins such as VP16 or 1CP4; as well as vaccinia or poxvirus inducible promoters.
The Examples below illustrate a generally applicable method that can be used to test putative promoters to readily determine whether or not they are helper-virus-inducible and whether or not they will be useful in the generation of high efficiency packaging cells. Briefly, the method involves replacing the p5 promoter of the AAV rep gene with the putative helper-virus-inducible promoter (either known in the art or identified using well-known techniques such as linkage to promoter-less "reporter" genes). The AAV rep-cap genes (with p5 replaced), preferably linked to a positive selectable marker such as an antibiotic resistance gene, are then stably integrated into a suitable host cell (such as the Hela or A549 cells exemplified below). Cells that are able to grow relatively well under selection conditions in the presence of the antibiotic) are then tested for their ability to express the rep and cap genes upon addition of a helper virus. As an initial test for rep and/or cap expression, cells can be readily screened using immunofluorescence to detect rep and/or cap proteins (as illustrated in the Examples below). Confirmation of packaging capabilities and efficiencies can then be determined by functional tests for replication and packaging of incoming rAAV vectors (also illustrated below). Using this methodology, we replaced the promoter with a helper-virus-inducible promoter derived from the mouse metallothionein gene, and used the resulting constructs to generate packaging cell lines capable of producing high titers of rAAV particles.
In our system, the AAV cap gene is also stably integrated into the packaging cell line. In a preferred embodiment, the rep and cap genes are introduced into the parental line together, by using a plasmid that contains them both (essentially as they are arranged in the AAV genome, except for replacement of the sequences upstream of rep, i.e. the promoter region). In illustrative examples below, we prepared WO 96/17947 PCT/US95/15892 28 a plasmid designated pMt-rep/cap//pKO-neo (shown in Figure 1).
The plasmid contains a heterologous promoter linked to a region containing the rep-cap genes. The rest of the rep-cap region, including the p19 promoter and the p40 promoter are retained. The plasmid also contains an AAV polyadenylation signal. Thus, the components of native AAV that are not present in the plasmid include the p5 promoter region (which has been substituted by the heterologous promoter) and the ITRs (which are present in the vector plasmid to be introduced separately).
Cells transfected with rep and cap genes as described above are then selected from untransfected cells according to methods that are routine in the art. Most conveniently, selection is accomplished by linking the rep and cap genes to one or more selectable markers (such as antibiotic resistance genes). For example, in the pMt-ret-rep/cap//pKO-neo construct described below, the neo-resistance gene was included next to the rep-cap sequences. Preferably, such selectable markers are driven by constitutive promoters; and preferably, such markers are introduced in an opposite orientation relative to the AAV rep-cap genes since that tends to reduce the potential for the promoter driving the selectable marker to effect expression of the rep gene (which can be detrimental to the host cell). After transfection and recovery, the cell lines are exposed to the antibiotic for which resistance has been provided (geneticin was used in the case of the constructs referred to above).
Integration into the host cell can be conveniently monitored using Southern analysis, for example. Expression of rep and cap proteins can be assayed using any of a variety of techniques; including structural assays (such as immunofluorescence), and functional assays (such as replication and packaging of an incoming rAAV vector) both of which are illustrated in the Examples below.
WO 96/17947 PCTIUS95/15892 29 In prior studies, the expression of rep protein in mammalian cells has been correlated with reductions in plating efficiencies and/or decreases in proliferation rates. For example, co-transfection of the rep coding sequence under the control of the metallothionein promoter and pSV2neo into human 293 cells resulted in the generation of neo-resistant cell lines which appeared to progress through DNA synthesis at a slower rate than control cells, even in the absence of heavymetals (see Yang et al. 1994 J. Virol. 68:4847-4856).
Since co-transfection requires the uptake and maintenance of both plasmids, but geneticin only selects for the neoresistance plasmid, the possibility exists that the repcontaining plasmid may be eventually lost. Moreover, if the introduction of rep exerts a negative effect on the host cell (even when the promoter is not induced), then there would be selective pressure favoring loss of rep activity. Based on those earlier observations, one would expect that cells that maintained and expressed rep would grow slowly. Conversely, cells could predictably resume normal growth rates by losing rep activity.
In our preferred system, the selectable marker is included on the same plasmid as the rep-cap sequences; and both are stably integrated into the host genome. .In the case of the plasmid Mt-rep/cap//pKOneo, for example, geneticinresistant cells would be expected to possess an integrated copy of the neo gene as well as pMt-rep/cap. Since the rep sequences cannot readily be lost in our system, the prior art would predict that the recipient cells would exhibit reduced growth rates.
Surprisingly, when our constructs were introduced into exemplary mammalian host cells (Hela and A549), the rate of proliferation of the geneticin-resistant clones was not significantly affected in either of the cell lines.
Based on the previously observed coupling between rep activity and reduced growth rate, one might expect that the cells exhibiting substantially normal growth rates represented WO 96/17947 PCTUS95/15892 30 hosts in which the rep gene had somehow become inactivated or lost.
In fact, additional experimentation (described below) indicated that the rep gene had been stably integrated into the resulting cell lines; and, moreover, that introduction of adenovirus into the medium resulted in the synthesis of relatively high levels of functional rep proteins.
These results suggested that our approach could be used to generate stable cell lines that were not subject to the sort of growth limitations observed in the past; and, as described below, subsequent experiments confirmed that these methods could be used to generate cell lines capable of efficiently packaging recombinant AAV vectors.
We have found, using these methods, that one can readily obtain packaging cells that are capable of replicating at least one half as rapidly as the parental cells, and capable of producing more than 100 rAAV particles/cell. In preferred embodiments of the present invention, the cells grow at least two-thirds as rapidly as the parental line, and produce more than 250 rAAV particles/cell. Using two different parental lines to date, we have generated packaging cells that replicate substantially as rapidly as the parent cells (at least about 80% of the rate), and that produce more'than about 500 rAAV particles per cell.
Generating rAAV vectors: To generate recombinant AAV particles useful for such purposes as gene therapy, the packaging cell line is supplied with a recombinant AAV vector comprising AAV inverted terminal repeat (ITR) regions surrounding one or more polynucleotides of interest (or "target" polynucleotides).
The target polynucleotide is operably linked to a promoter, either its own or a heterologous promoter. A large number of suitable promoters are known in the art, the choice of which depends on the desired level of expression of the target polynucleotide; whether one wants constitutive WO 96/17947 PCT/US9/15892 31 expression, inducible expression, cell-specific or tissuespecific expression, etc.
Preferably, the rAAV vector will also contain a positive selectable marker in order to allow for selection of cells that have been infected by the rAAV vector. Negative selectable markers can also be included; as a means of selecting against those same cells should that become necessary or desirable. In a preferred embodiment, one can make use of the "bifunctional selectable fusion genes" described by S.D. Lupton; see, PCT/US91/08442 and PCT/US94/05601. Briefly, those constructs involve direct translational fusions between a dominant positive selectable marker a negative selectable marker. Preferred positive selectable markers are derived from genes selected from the group consisting of hph, neo, and gpt, and preferred negative selectable markers are derived from genes selected from the group consisting of cytosine deaminase, HSV-I TK, VZV TK, HPRT, APRT and gpt. Especially preferred markers are bifunctional selectable fusion genes wherein the positive selectable marker is derived from hph or neo, and the negative selectable marker is derived from cytosine deaminase or a TK gene.
By way of illustration, we have used rAAV vectors containing polynucleotides that encode a functional cystic fibrosis transmembrane conductance regulator polypeptide (CFTR) operably linked to a promoter. As is now known in the art, there are a variety of CFTR polypeptides that are capable of reconstructing CFTR functional deficiencies in cells derived from cystic fibrosis patients. For example, Rich et al. (1991, Science, 253: 205-207) described a CFTR derivative missing amino acid residues 708-835, that was capable of transporting chloride and capable of correcting a naturally occurring CFTR defect. Egan et al. (1993) described another CFTR derivative (comprising about 25 amino acids from an unrelated protein followed by the sequence of native CFTR beginning at residue 119) that was also capable of restoring WO 96/17947 PCT/US95/15892 32 electrophysiological characteristics of normal CFTR. To take two additional examples, Arispe et al. (1992, Proc. Natl.
Acad. Sci. USA 89: 1539-1543) showed that a CFTR fragment comprising residues 433-586 was sufficient to reconstitute a correct chloride channel in lipid bilayers; and Sheppard et al. (1994, Cell 76: 1091-1098) showed that a CFTR polypeptide truncated at residue 836 to about half its length was still capable of building a regulated chloride channel. Thus, the native CFTR protein, and mutants and fragments thereof, all constitute CFTR polypeptides that are useful under this invention.
Other useful target polynucleotides can be used in this invention to generate rAAV vectors for a number of different applications. Such polynucleotides include, but are not limited to: polynucleotides encoding proteins useful in other forms of gene therapy to relieve deficiencies caused by missing, defective or sub-optimal levels of a structural protein or enzyme; (ii) polynucleotides that are transcribed into anti-sense molecules; (iii) polynucleotides that are transcribed into decoys that bind transcription or translation factors; (iv) polynucleotides that encode cellular modulators such as cytokines; polynucleotides that can make recipient cells susceptible to specific drugs, such as the herpes virus thymidine kinase gene; and (vi) polynucleotides for cancer therapy, such as the wild-type p53 tumor suppressor cDNA for replacement of the missing or damaged p53 gene associated with some lung and breast cancers.
Since the therapeutic specificity of the resulting recombinant AAV vector is determined by the plasmid introduced, the same packaging cell line can be used for any of these applications. The plasmid comprising the specific target polynucleotide is introduced into the packaging cell for production of the AAV vector by one of several possible methods; including, for example, electroporation.
Helper virus can be introduced before, during or after introduction of the rAAV vector. As illustrated in Example WO 96/17947 PCTIUS95/15892 33 the plasmid can be co-infected into the culture along with the helper virus. The cells are then cultured for a suitable period, typically 2-5 days, in conditions suitable for replication and packaging as known in the art (see references above and examples below). Lysates are prepared, and the recombinant AAV vector particles are purified by techniques known in the art.
In a preferred embodiment, also illustrated in the Examples below, the recombinant AAV vector is itself stably integrated into a clone of the packaging cell line. Such a stable, vector-containing packaging line can be grown and stored until ready for use. To induce production of rAAV particles, the user simply infects the cells with helper virus and cultures the cells under conditions suitable for replication and packaging of AAV (as described below).
Using the cell lines for assaying AAV titer: We have also used the packaging cell lines of the present invention to set up an rAAV infectious titer assay.
In particular, as described below, we have demonstrated that there is a linear relationship between incoming rAAV and replicated rAAV over a range of greater than two logs.
Thus, in another embodiment of this invention, -we provide a method for determining the relative infectious titer of an rAAV preparation. Although the amount of helper virus and the incubation time influence the amount of rep activity, they can be readily optimized and kept constant, as illustrated below.
To conduct the assay, aliquots of the packaging cell line are introduced with a standard amount of helper virus and serial dilutions of the rAAV preparation to be tested. The relative infectious titer of the AAV is indicated by the amount of replicated AAV present in each aliquot after suitable incubation; and can be compared to a preparation of known titer.
WO 96/17947 WO177 PCT/US95/15892 34 The examples presented below are provided as a further guide to the practitioner of ordinary skill in the art, and are not to be construed as limiting the invention in any way.
EXAMPLES
Example 1 Construction of a plasmid encoding the rep-cap sequences operably linked to a heterologous promoter As a first illustration, we constructed a plasmid containing the wild type rep and cap genes (from deoxyribonucleotide 311 to 4493 of the AAV genome); operably linked to the mouse metallothionein I (mMt-I) gene promoter (from -650 to +64 relative to the start of transcription; as described by Srivastava et al. 1983. J. Virol. 45:555-564; and Hamer and Walling (1992) J. Mol. Appl. Genet. 1:273-288).
This construction effectively removes both ITR's and substitutes the mMt-I promoter for the p5 promoter while maintaining all of the AAV reading frames, the pl9 and promoters and the polyadenylation signal. Also included within this vector is pKOneo, which contains the neo gene {providing resistance to neomycin and gentamicin) under control of the SV40 early promoter; as well as SV40 small t intron and polyadenylation signal oriented in the opposite transcriptional direction relative to pMt-rep/cap (Ito et al.
1994 Cancer Lett. 76:33-39).
Standard procedures were followed for plasmid construction, growth and purification (Ausubel et al. (ed.) 1987. Current Protocols in Molecular Biology, Greene Publishing Associates, Brooklyn, The resulting plasmid, designated pMt-rep/cap//pKo-neo, is shown in Figure 1.
WO 96/17947 PCTIUS95/15892 35 Example 2 Integration of the rep-cap genes into mammalian cell lines We introduced plasmid pMt-rep/cap//pKO-neo into exemplary mammalian cell lines in order to examine expression of the rep and cap genes after integration into the host cell genome.
Human Hela and A549 [American Type Culture Collection (ATCC)] cells were grown in Dulbecco's modified Eagle's medium (DMEM) supplemented with antibiotics and 10% fetal bovine serum. All DNA transfections were carried out by electroporation of 15-30 pg pMt-rep/cap//pKO-neo plasmid into 4 X 106 cells in 800 Ml serum-free medium in a 0.4 cm cuvette at 250 volts/960 gF using a Gene Pulser (Bio-Rad).
After electroporation, the cells were plated at low density in the presence of 1 mg/ml active component geneticin (Gibco-BRL). Individual colonies were ring cloned, expanded and maintained in 1 mg/ml geneticin.
In prior studies, the expression of rep protein in mammalian cells has been correlated with reductions in plating efficiencies and/or decreases in proliferation rates. For example, co-transfection of the rep coding sequence under the control of the metallothionein promoter and pSV2neo into human 293 cells resulted in the generation of neo-resistant cell lines which appeared to progress through DNA synthesis at a slower rate than control cells, even in the absence of heavymetals (see Yang et al. 1994 J. Virol. 68:4847-4856).
Since co-transfection requires the uptake and maintenance of both plasmids, but geneticin only selects for the neoresistance plasmid, the possibility exists that the repcontaining plasmid may be eventually lost. Moreover, if the introduction of rep exerts a negative effect on the host cell (even when the promoter is not induced), then there would be selective pressure favoring loss of rep activity. Based on those earlier observations, one would expect that cells that maintained and expressed rep would grow slowly. Conversely, WO 96/17947 PCTIUS95/15892 36 cells could predictably resume normal growth rates by losing rep activity.
In our preferred system, the selectable marker is included on the same plasmid as the rep-cap sequences; and both are stably integrated into the host genome. In the case of the plasmid Mt-rep/cap//pKOneo, for example, geneticinresistant cells would be expected to possess an integrated copy of the neo gene as well as pMt-rep/cap. Since the rep sequences cannot readily be lost in our system, the prior art would predict that the recipient cells would exhibit reduced growth rates.
Surprisingly, when our constructs were introduced into exemplary mammalian host cells (Hela and A549), the rate of proliferation of the geneticin-resistant clones was not significantly affected in either of the cell lines.
Based on the previously observed coupling between rep activity and reduced growth rate, one might expect that the cells exhibiting substantially normal growth rates represented hosts in which the rep gene had somehow become inactivated or lost.
In fact, additional experimentation (described below) indicated that the rep gene had been stably integrated into the resulting cell lines; and, moreover, that introduction of adenovirus into the medium resulted in the synthesis of relatively high levels of functional rep proteins.
These results suggested that our approach could be used to generate stable cell lines that were not subject to the sort of growth limitations observed in the past; and, as described below, subsequent experiments confirmed that these methods could be used to generate cell lines capable of efficiently packaging recombinant AAV vectors.
By combining the methodology described herein with the teachings and materials available in the art, one of ordinary skill in the field will readily be able to prepare embodiments of the present invention such as those illustrated in these Examples. For convenience, however, we are depositing an 37 exemplary plasmid (pMt-rep/cap//pKO-neo), as well as exemplary packaging cells (Hela clone 37 and A549 clone 20) (all as described in these Examples) with the American Type Culture Collection, 12301 Parklawn Dr., Rockville, Maryland 29852, having Accession Numbers 97049, CRL-11831 and CRL-11877, respectively.
The deposits were made under the terms of the Budapest Treaty and are incorporated herein by reference. Upon allowance and issuance of this application as a United States Patent, all restrictions on availability of the deposit will be Go .10 irrevocably removed; and access to the designated deposits S. will be available during pendency of the above-named application to one determined by the Commissioner to be entitled thereto under 37 CFR 1.14 and 35 USC 1.22. Moreover, the designated deposits will be maintained for a period of thirty years from the date of deposit, or for five years after the last request for the deposit; or for the enforceable life of the U.S. patent, whichever is longer. The deposited materials mentioned herein are intended for convenience only, and are not required to practice the present invention in view of the descriptions herein; and in addition these materials are incorporated herein by reference.
Example 3 Detection of Rep protein by indirect immunofluorescence We examined rep protein expression in the transfected cell lines (exemplary clones derived from Hela or A549 cells) using indirect immunofluorescence (IF).
Transfected and control cells were seeded (at 1.6 x 104 cells/chamber) onto Lab-Tek 8 well chamber slides (Nunc Inc., Ill.) in 200 pl culture medium and incubated overnight. The cells were then treated individually or in combination with wild type AAV (MOI=5), adenovirus (MOI=10) and ZnS04 (100 AM) for -30 hours. After removing the culture medium, the cells were washed three times with phosphate buffered saline (PBS) at 0 C and fixed for 1-2 min. with acetone. The cells were then washed three additional times with PBS and incubated WO 96/17947 PC1US95/15892 38 overnight with "WT" medium nonfat dry milk, 0.5 mg/ml bovine serum albumin, 150 mM NaCl, 50 mM HEPES (pH 0.1% Tween 20 and 1 mM NaN 3 Anti-rep antibody (rabbit anti-Rep78.93; Trempe et al.
1987 Virology 161:18-28) was diluted 1:250 in WT and 100 gl added to each well for 1 hour at room temperature The cells were washed five times with WT and then incubated with 100 gl of a 1:100 dilution of anti-rabbit IgG FITC conjugate secondary antibody (Sigma Chemical Corp.) in the dark for 1 hour at RT. The cells were then washed three times with WT and two times with PBS in the dark and examined with an Axioskop H fluorescence microscope (Zeiss, Germany).
When adenovirus was included in the culture medium, rep protein was detectable in a number of the cells examined (8 out of 23 A549 clones and 3 out of 28 Hela clones). The addition of heavy metals did not significantly affect the observed rep expression under any conditions.
These observations suggest that very little, if any, rep protein is synthesized in these cells in the absence of an adenovirus infection. Southern analysis of genomic DNA isolated from a subset of the rep (IF) positive cells hybridized with a labeled cap probe demonstrated that a single copy of the rep-cap sequences was integrated into the cellular
DNA.
Although not wishing to be bound by theory regarding the mechanism by which the metallothionein promoter is helpervirus-inducible, our results suggest that it may contain an adeno-responsive element, which would be consistent with an observation made by Carthew et al., 1985, Cell 43:439-448.
In any case, the method we describe for obtaining and testing a helper-virus-inducible promoter is a general one that can be readily applied to any promoter of potential interest by simply swapping it into rep constructs and screening for colonies as we describe herein.
WO 96/17947 PCT/US95/15892 39 In subsequent experiments, exemplary clones (of Hela and A549 origin) were tested for their ability to replicate recombinant AAV genomes after infection, as described below.
Example 4 Replication activity of IF+ cells We examined whether the pMt-rep/cap//pKO-neo transfected cell lines exhibited functional replication activity. We selected a recombinant AAV preparation including the cystic fibrosis cDNA (AAVCFTR) (Riordan et al. 1989 Science 245:1066- 1073) as an exemplary recombinant AAV. Serial dilutions of the rAAV preparation were incubated adenovirus (MOI=25) on rep IF+ cells (Hela clone 37 and A549 clone 20). AAVCFTR virus was diluted from 1.2 x 108 to 1.2 x 103 particles (as determined by slot blot) and incubated on both cell lines x 10 5 cells/well in a 6 well dish) for 48 hours adenovirus (MOI 25 pfu/cell). In order to control for the effect of adenovirus infection on the cells infected with the virus, the culture medium from each well was removed to a labeled tube and any cells still attached to the culture dish were trypsinized and pooled with cells present in the media. The cell suspension was centrifuged at 3000 rpm for 5 min., after which the supernatant was removed and total nucleic'acid was prepared from the cell pellet (according to Ausubel et al.
1987 Current Protocols in Molecular Biology Greene Publishing Associates, Brooklyn, Negative controls for the experiment included the incubation of 1.2 x 108 AAVCFTR particles on either cell line without adenovirus.
Fifteen micrograms of nucleic acid for each sample, as well as untreated Hela clone 37 DNA 20 pg of AAVCFTR plasmid (positive control for Southern), was digested with EcoRI, subjected to gel electrophoresis, transferred to nitrocellulose and probed with a 1.488 kb EcoRI fragment from within the CFTR cDNA. Lanes 15-18 (Fig. 2) contain parental Hela and A549 genomic DNA 20 pg AAVCFTR plasmid to show WO 96/17947 PCT/US95/15892 40 the hybridization pattern of the endogenous human CFTR gene (Rommens et al. 1989 Science 245:1059-1065) and the 1.488 kb CFTR cDNA signal, respectively. A hybridization signal migrating at 1.488 kb is present in DNA isolated from both the Hela clone 37 and A549 clone 20 cell lines after infection by AAVCFTR virus and adenovirus (Fig. 2, lanes 1, 2 and 8).
These results demonstrated that both Hela clone 37 and the A549 clone 20 lines possessed sufficient rep activity to replicate the incoming AAVCFTR DNA upon infection, although both lines absolutely required adenovirus (Fig. 2, compare lanes 1 and 7 or 8 and 14).
Example rAAV infectious titer assays Additional rep activity assays were performed in order to determine whether there was a linear relationship between incoming AAVCFTR virus and replicated AAVCFTR sequences (which could be exploited as the basis of an rAAV infectious titer assay).
Three log dilutions from 1.2 x 109 to 1.2 x 107 AAVCFTR particles, as determined by slot blot hybridization, were cultured in 2.5 ml media on 2.5 x 10 5 Hela clone 37 cells plus adenovirus (MOI 25 pfu/cell) for 48 hours in a 6 well culture dish. As a negative control, 1.2 x 109 AAVCFTR particles minus adenovirus was included. After 48 hours, total nucleic acid was prepared from the cells for Southern analysis to detect replicated CFTR sequences, as previously described (Fig. Hela clone 37 cells cultured in the presence of 1.2 x 109 AAVCFTR particles only (Fig. 3, lane 1) displayed a hybridization pattern reflecting the genomic organization of the endogenous human CFTR gene. The addition of 20 pg of AAVCFTR plasmid into Hela clone 37 DNA shows where the 1.488 kb EcoRI fragment migrates on this blot (Fig. 3, lane The samples from the serial dilutions of AAVCFTR virus plus adenovirus are shown in lanes 2-4, respectively (Fig. The hybridization observed at 1.488 kb appears to WO 96/17947 PCT/US95/15892 41 be proportional to the input AAVCFTR virus and absolutely dependent on the presence of adenovirus (compare lanes 1 and 2).
These results demonstrate that packaging cells produced according to the present invention are capable of replicating an AAV genome in the presence of adenovirus, and that this activity can be used to quantify the number of infectious viral particles present in a given sample. In this particular assay, the linear response required at least 1.2 x 107 AAVCFTR particles cultured on 2.5 x 105 Hela clone 37 cells MOI 48 particles/cell). The particle number was determined by slot blot hybridization of the AAVCFTR virus preparation and may reflect the contribution of infectious and defective AAVCFTR particles; whereas the infectious assay described above should only detect infectious particles.
Example 6 AAVCFTR plasmid DNA gene transfer into packaing cells Current methods for the production of recombinant AAV (rAAV) virus include the transient transfection of plasmid vectors containing the rAAV sequences. One or more steps are undertaken to remove the plasmid DNA from a rAAV preparation.
AAVCFTR plasmid DNA was incubated directly onto packaging cells (Hela clone 37) adenovirus to determine whether the above-described infection assay would detect non-viral DNA.
As a control, AAVCFTR plasmid (10 pg) was electroporated as previously described into 4 x 106 Hela clone 37 cells and then transferred to a 100 mm culture dish. The following day, the cells were washed in phosphate buffered saline (PBS), trypsinized and 2.5 x 105 cells seeded in duplicate in a 6-well dish adenovirus (MOI 25 pfu/cell). Seven additional wells were seeded with 2.5 x 105 Hela clone 37 cells each and cultured with a dilution series of AAVCFTR plasmid adenovirus (MOI 25 pfu/cell) or AAVCFTR virus (1 x 109 particles) plus adenovirus. Total nucleic acid was prepared WO 96/17947 PCTIUS95/15892 42 48 hours post-infection and Southern analysis was performed as previously described. Lanes 9 and 10 (Fig. 4) show the hybridization pattern of the endogenous CFTR gene and the migration of CFTR cDNA (20 pg) spiked into human genomic DNA when digested with EcoRI and probed with the 1.488 kb CFTR cDNA fragment. Electroporation of the AAVCFTR plasmid into Hela clone 37 cells resulted in a signal migrating at 1.488 kb (Fig.4, lane 1) and represents the amount of AAVCFTR plasmid present in these cells 24 hours after transfection. An equal number of electroporated cells incubated with adenovirus reveals significantly more signal migrating at 1.488 kb reflecting the ability of Hela clone 37 cells to amplify the AAVCFTR plasmid genome after electroporation (Fig. 4, lane 2).
Lanes 3 and 4 show the result of incubating 5 Ag AAVCFTR plasmid adenovirus, respectively) directly onto Hela clone 37 cells without transfection. Although there is a small amount of hybridization which is amplified in the presence of adenovirus, the signal is significantly lower than that derived from either AAVCFTR plasmid transfection (Fig. 4, lane 2) or AAVCFTR virus infection (Fig. 4, lane 11,).
Lanes 5-8 show the results of incubating 1 gg, 100 ng, 10 ng and 1 ng AAVCFTR plasmid, respectively, on Hela clone 37 cells in the presence of adenovirus.
These results demonstrate that, in the presence of adenovirus infection, the packaging cells produced according to the present invention express rep protein and are able to replicate recombinant AAV plasmid DNA genomes introduced by transfection. This activity was dependent upon the transfection of AAV plasmid DNA into the packaging cells (e.g.
co-incubation of plasmid DNA containing AAV sequences and adenoviral infection did not result in significant replication of the input plasmid).
WO 96/17947 PCT/US95/15892 43 Example 7 Time course of rAAV infectious titer assay A time course examining the replication of AAVCFTR virus after infection of packaging cells in the presence of adenovirus was performed to verify that the time of viral incubation was not influencing the rAAV infectious assay described above.
To determine the time course of AAVCFTR virus replication, 1.5 x 105 Hela clone 37 cells were cultured with 1.2 x 109 AAVCFTR particles and adenovirus (MOI 5 pfu/cell) in a 6-well dish and harvested at various times after infection for Southern analysis. Control human DNA spiked AAVCFTR plasmid (20 pg) was also included in the Southern as a control (Fig. 5, lanes 12, 13). Lanes 2-6 show the result of harvesting total nucleic acid at 20, 24, 39, 42 and 48 hours post-infection (Fig. The signal observed at 1.488 kb increases to a maximum by 39 hours (Fig. 5, lane 4) and a comparison of lanes 1 and 6 demonstrates the dependence of this assay on input adenovirus.
These results demonstrate that the infectious rAAV titering assay described in Example 5 had proceeded for a sufficient amount of time to reach a maximum.
Example 8 Adenovirus titration of rAAV infectious titer assay The effect of adenovirus MOI on the rAAV infectious titer assay was performed to confirm that adenovirus was not limiting in the assay.
The dilution of input adenovirus from MOI=25.0 to 0.1 cultured with 1.2 x 109 AAVCFTR particles on 2.5 x 105 Hela clone 37 cells and harvested at 48 hours did not reduce the signal intensity migrating at 1.488 kb (Fig. 5, lanes 7-11).
These data, taken together, show that neither the time of incubation nor the amount of added adenovirus significantly impacts the initial titering results presented in Examples 4 and WO 96/17947 PCTIUS95/15892 44 Example 9 Production of infectious AAVCFTR virions after transfection We then demonstrated that the AAVCFTR plasmid, introduced into packaging cells of the present invention, could be effectively packaged into infectious AAVCFTR virions.
In particular, AAVCFTR plasmid (10 pg) was electroporated as previously described into 2 x 106 exemplary packaging cells (Hela clone 37) and plated onto a 100 mm culture dish. The following day the cells were washed with PBS, trypsinized, pelleted and brought up in culture media. The cells were counted and 2 x 105 seeded into two wells of a 6-well culture dish. The cells were incubated with 2.5 ml culture media adenovirus (MOI=10). After 48 hours, the cells were scraped into the media and subjected to 3 freeze/thaw cycles in dry ice/EtOH bath and 370C H20 bath, respectively. After centrifugation at 3000 rpm for 10 min., the supernatant was removed ml) for titering and total nucleic acid was isolated from the post freeze/thaw cell debris pellets to look for rAAVCFTR replicative intermediates. The supernatant from the electroporated packaging cells plus adenovirus (50 and 500 Al) and one dilution (500 pl) of the non-adenovirus infected control were cultured in a 2.5 ml final volume for 48 hours in the presence of additional adenovirus (MOI5) for titering as described in Examples 4 and 5. No steps were undertaken to remove potential plasmid contamination from the electroporation or adenovirus from the initial cultures to generate additional rAAVCFTR virions.
Southern analysis of nucleic acid isolated from the post freeze/thaw cell pellets, digested with EcoRI and probed for CFTR sequences is shown in Figure 6. As described previously in Figure 4, AAVCFTR plasmid can be detected in Hela clone 37 cells after electroporation, and the hybridization signal migrating at 1.488 kb increases after infection with adenovirus (Fig. 6, lanes 1 and 2).
The titering of supernatants derived from Hela clone 37 cells electroporated with AAVCFTR plasmid adenovirus is WO 96/17947 PCT/US95/15892 45 presented in Figure 7, lanes 1-3. A slight signal migrating at 1.488 kb can be detected in DNA isolated from Hela clone 37 cells incubated with supernatant derived from the minus adenovirus control and reflects a small amount of contaminating input AAVCFTR plasmid from the electroporation (Fig. 7, lane Supernatant derived from a duplicate well cultured with adenovirus and titered on Hela clone 37 cells revealed significantly more hybridization migrating at 1.488 kb relative to control conditions (Fig. 7, compare lanes 3 and i).
These results demonstrate that after transfection of packaging cells with a recombinant AAV plasmid and infection with adenovirus, the rAAV sequences were replicated and assembled into infectious rAAV virions.
Example Production of infectious recombinant AAV virions after infection with rAAV virus We further examined the ability of the packaging cells to generate infectious rAAV virions after infection. Several control experiments were included to determine whether newly synthesized virus was generated. These include the use of parental Hela cells and conditions which should not allow for the generation of rAAV. Both Hela clone 37 and parental Hela cells (2.5 x 105) were seeded onto 6-well dishes and incubated for 48 hours under the following conditions: plus 109 AAVCFTR particles alone; plus 109 AAVCFTR particles with either adenovirus (MOI=10) or wild type AAV (MOI=5); or all three.
After 48 hours, the cells were harvested as described in Example 9. Nucleic acid was isolated from the post freeze/thaw cell pellets for Southern analysis and the supernatants titered.
Southern analysis of nucleic acid isolated from either Hela clone 37 cells (lanes 3-6) or parental Hela cells (lanes 7-10) treated under the conditions described above is presented in Figure 6. Treatment of either cell line with 109 WO 96/17947 PCT/US95/15892 46 AAVCFTR particles alone did not result in the generation of replicated AAVCFTR sequences (Fig. 6, lanes 3 and 7, respectively). The addition of adenovirus to Hela clone 37 cells, but not to parental Hela cells, resulted in the generation of a significant amount of signal migrating at 1.488 kb, consistent with previous observations (Fig. 6, lanes 4 and Wild-type AAV did not result in the generation of replicated AAVCFTR in either cell line, due to the lack of adenovirus helper function (Fig. 6, lanes 5 and Finally, the co-infection of 10 9 AAVCFTR particles with both adenovirus and wild type AAV results in the generation of replicated AAVCFTR sequences in both Hela clone 37 and parental Hela cells (Fig. 6, lanes 6 and 10). The signal migrating at 1.488 kb may be reduced in the Hela clone 37 cells and the overall lane background is increased due to competition between the wild type and recombinant AAVCFTR virus replication.
The supernatants generated after freeze/thawing were incubated on fresh Hela clone 37 cells (500 gl diluted to ml unless otherwise indicated) in the presence of adenovirus (MOI=5) for titering. No steps were undertaken to remove input AAVCFTR virus, wild type AAV or adenovirus from the initial cultures to generate additional rAAVCFTR virions.
After 48 hours, total nucleic acid was prepared and'Southern analysis performed as previously described for the detection of AAVCFTR replication.
The result of culturing both Hela clone 37 and parental Hela cells with 109 AAVCFTR particles under various conditions is presented in Figure 7, lanes 4-12. All lanes reveal the presence of rAAVCFTR virus in the supernatants which is due to the fact that no attempt was made to purify de novo generated virions from input virus. Any signal above the input signal should correlate with conditions that resulted in the amplification of AAVCFTR signal observed in Figure 6. These conditions include: 109 AAVCFTR particles plus adenovirus and/or wild type AAV in parental Hela cells (Fig. 6, lanes 4, 6 and 10). All other conditions did not result in the WO 96/17947 PCT/US95/15892 47 replication of AAVCFTR sequences and should display a similar signal after titering, reflecting the presence of input AAVCFTR virus (Fig. 7; lanes 4, 7, 9-11).
As expected, co-incubation of AAVCFTR, adenovirus and wild type AAV resulted in the generation of de novo AAVCFTR virus production in both Hela clone 37 and parental Hela cells (Fig. 7; lanes 8 and 12). AAVCFTR virus production was also observed in Hela clone 37 cells, but not in parental Hela cells, with adenovirus alone (Fig. 7, compare lane 6 and 4), consistent with the observed increase in AAVCFTR signal in DNA isolated from these cells in Figure 6, lane 4. The difference in intensity of the hybridization observed at 1.488 kb in lane 6 (Fig. 7) versus the signal representing input AAVCFTR virus (lanes 4, 7, 9-11) reveals the increase in AAVCFTR titer upon the infection and amplification of AAVCFTR in Hela clone 37 cells.
These results demonstrate that packaging cells, in the presence of adenovirus, can replicate and package rAAV vector genomes into infectious virions by either transfection or infection (Figures 6 and 7).
Example 11 Rescue and amplification of an integrated rAAV vector from packaing cells Packaging cells (derived from Hela clone 37) were electroporated as previously described with a recombinant AAV vector (designated rAAV-CMV-Hygro) containing the hygromycin resistance gene operably linked to the CMV enhancer/promoter; and a stable, polyclonal line was derived by selection in 300 Mg/ml hygromycin.
In order to determine whether the rep activity present in the packaging cells could effectively recognize, excise and amplify the integrated rAAV-CMV-Hygro vector, the polyclonal, hygro-resistant Hela clone 37 line (2.5 x 105 cells/well) was seeded onto a 6 well dish for infection with adenovirus alone, plus wild type AAV (MOI=5) or uninfected.
WO 96/17947 PCTIUS95/15892 48 After 48 hours, nucleic acid was prepared, as previously described, digested with NheI/Asp718 and Southern analysis performed using a labeled, 1048 bp NheI/Asp718 fragment containing the hygromycin resistance gene as the probe.
Figure 8 shows the result of this analysis. Lane 1 represents DNA isolated from the parental Hela clone 37 cell line, and hence does not contain the hygro-resistance gene. Lane 2 contains DNA from the polyclonal, hygro-resistant Hela clone 37 line which at this exposure time does not show the presence of the resistance gene which is present at an average of about 1 copy/well (data not shown). DNA isolated from a duplicate well containing the hygro-resistant Hela clone 37 cells treated with adenovirus was run in lane 3. The hybridization present at 1048 bp represents material derived from the excision and amplification of the integrated AAVCMVHygro vector integrated in the Hela clone cells. The addition of wild-type AAV to the adenovirus infection gave similar results (lane 4).
These results demonstrate that the rep activity generated in the rAAV packaging cells is sufficient to recognize, excise and amplify an integrated recombinant AAV vector.
48/1 Deposit Details Exemplary plasmid and exemplary packaging cells as described in the above examples were deposited at the American Type Culture Collection with the following details: Sample plasmid pMt-rep/cap//pKo-neo Cell HeLa clone 37 Cell A549 clone 20 Accession Number 97049 CRL-11831 CRL-11877 Date of Deposit 10 February 1995 10 February 1995 19 April 1995 The deposits were made and referred to herein as a matter of convenience and are not required to practice the present invention in view of the descriptions provided herein.
.0 0 a 0 00 0 0 0 000 0
S

Claims (19)

1. A method of producing a mammalian cell for packaging of a recombinant AAV (rAAV) vector, said method including the steps of: providing a mammalian cell which includes a stably integrated AAV cap gene operably linked to a promoter, and a stably integrated AAV rep gene operably linked to a helper-virus inducible heterologous promoter, wherein p5 promoter function has been replaced by the helper-virus inducible heterologous promoter; replicating the cell of step to produce a population of cells; and introducing a helper virus to the population of cells of step 15 wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep gene.
2. A method according to claim 1, wherein said helper virus is an adenovirus.
3. A method according to claim 1, wherein said packaging cell grows at least one half as rapidly as parental-type cells that do not contain an AAV rep •gene, and wherein said packaging cell when used to package rAAV vectors produces at least 100 rAAV particles/cell.
4. A method according to claim 1, wherein said mammalian cell of step includes the combined rep and cap genes of AAV in which the promoter has been replaced by a heterologous promoter.
5. A method according to claim 4, wherein said heterologous promoter is a mouse metallothionein I (mMT-I) promoter.
6. A cell produced by the method of claim 1, and progeny thereof, wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep gene.
7. A cell produced by the method of claim 3, and progeny thereof, wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep gene.
8. A cell produced by the method of claim 4, and progeny thereof, wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep gene.
9. A cell produced by the method of claim 5, and progeny thereof, wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep gene.
A mammalian cell for packaging of a recombinant AAV (rAAV) vector, said cell including a stably integrated cap gene operably linked to a promoter, i: 15 and a stably integrated rep gene operably linked to a helper virus-inducible heterologous promoter; wherein p5 promoter function has been replaced by the helper virus-inducible heterologous promoter and wherein said cell exhibits helper virus-inducible expression of said stably integrated AAV rep *".gene.
11. An AAV packaging cell of claim 10, wherein said helper-virus- inducible expression of said stably integrated AAV rep gene is inducible by adenovirus.
12. An AAV packaging cell of claim 10, wherein said packaging cell grows at least one half as rapidly as parental-type cells that do not contain an AAV rep gene, and wherein said packaging cell when used to package rAAV vectors produces at least 100 rAAV particles/cell.
13. An AAV packaging cell of claim 10, wherein said cell includes the combined rep and cap genes of AAV in which the p5 promoter has been replaced by a heterologous promoter.
14. An AAV packaging cell of claim 13, wherein said heterologous promoter is a mouse metallothionein I (mMT-I) promoter.
An AAV packaging cell of claim 10, further including a stably integrated recombinant AAV (rAAV) vector, said vector including a polynucleotide sequence of interest located between two AAV inverted terminal repeat (ITR) regions, wherein said polynucleotide is operably linked to a promoter.
16. A method of packaging a recombinant AAV vector, including the steps of: providing an AAV packaging cell of claim introducing a recombinant AAV (rAAV) vector, said vector including a polynucleotide sequence of interest located between two AAV inverted terminal repeat (ITR) regions, wherein said polynucleotide is operably linked to a promoter; S* introducing a helper virus; and incubating the cell under conditions suitable for replication and packaging of AAV such that said rAAV vector is packaged.
17. A method of packaging a recombinant AAV vector, including the steps of: providing an AAV packaging cell of claim 15 which includes a stably integrated rAAV vector including a polynucleotide of interest operably linked to a promoter; introducing a helper virus; and incubating the cell under conditions suitable for replication and packaging of AAV such that said rAAV vector is packaged.
18. A method of determining the relative infectious titer of an rAAV vector preparation, including the steps of: introducing a helper virus and serial dilutions of the rAAV vector preparation to AAV packaging cells of claim incubating the cells under conditions suitable for replication of AAV; and determining the amount of replicated rAAV vector relative to an rAAV preparation of known titer.
19. The method of claim 1, further including the step of selecting a cell exhibiting helper-virus-inducible expression of said stably integrated AAV rep gene. Dated this nineteeth day of May 1999. TARGETED GENETICS CORPORATION Patent Attorneys for the Applicant: F B RICE CO *t ft
AU45963/96A 1994-12-06 1995-12-06 Packaging cell lines for generation of high titers of recombinant AAV vectors Ceased AU707866B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US35021994A 1994-12-06 1994-12-06
US08/350219 1994-12-06
US48057595A 1995-06-07 1995-06-07
US08/480575 1995-06-07
PCT/US1995/015892 WO1996017947A1 (en) 1994-12-06 1995-12-06 Packaging cell lines for generation of high titers of recombinant aav vectors

Publications (2)

Publication Number Publication Date
AU4596396A AU4596396A (en) 1996-06-26
AU707866B2 true AU707866B2 (en) 1999-07-22

Family

ID=26996529

Family Applications (1)

Application Number Title Priority Date Filing Date
AU45963/96A Ceased AU707866B2 (en) 1994-12-06 1995-12-06 Packaging cell lines for generation of high titers of recombinant AAV vectors

Country Status (5)

Country Link
EP (1) EP0796339A1 (en)
JP (1) JPH10511264A (en)
AU (1) AU707866B2 (en)
CA (1) CA2207927A1 (en)
WO (1) WO1996017947A1 (en)

Families Citing this family (265)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6326356B1 (en) 1996-10-18 2001-12-04 Board Of Regents, The University Of Texas System Suppression of neu overexpression using a mini-E1A gene
US6924128B2 (en) 1994-12-06 2005-08-02 Targeted Genetics Corporation Packaging cell lines for generation of high titers of recombinant AAV vectors
US5843742A (en) * 1994-12-16 1998-12-01 Avigen Incorporated Adeno-associated derived vector systems for gene delivery and integration into target cells
US20020103147A1 (en) 1997-09-05 2002-08-01 Hammond H. Kirk Gene therapy for congestive heart failure
US6306830B1 (en) 1996-09-05 2001-10-23 The Regents Of The University Of California Gene therapy for congestive heart failure
CA2188575A1 (en) 1995-02-28 1996-09-06 H. Kirk Hammond Gene transfer-mediated angiogenesis therapy
US6752987B1 (en) 1995-02-28 2004-06-22 The Regents Of The University Of California Adenovirus encoding human adenylylcyclase (AC) VI
DE19625188A1 (en) * 1996-06-24 1998-01-08 Medigene Gmbh System for the production of AAV vectors
US6294370B1 (en) 1997-06-24 2001-09-25 Medigene Ag System for the production of AAV vectors
US6541012B2 (en) 1996-06-24 2003-04-01 Christoph Bogedain System for the production of AAV vectors
US5866552A (en) * 1996-09-06 1999-02-02 The Trustees Of The University Of Pennsylvania Method for expressing a gene in the absence of an immune response
WO1998010088A1 (en) * 1996-09-06 1998-03-12 Trustees Of The University Of Pennsylvania An inducible method for production of recombinant adeno-associated viruses utilizing t7 polymerase
EP0950111A1 (en) 1996-09-06 1999-10-20 The Trustees Of The University Of Pennsylvania Methods using cre-lox for production of recombinant adeno-associated viruses
JP2001506133A (en) 1996-12-18 2001-05-15 ターゲティッド ジェネティクス コーポレイション AAV split-packaging genes and cell lines containing such genes for use in producing recombinant AAV vectors
JP2001506132A (en) * 1996-12-18 2001-05-15 ターゲティッド ジェネティクス コーポレイション Recombinase activatable AAV packaging cassette for use in producing AAV vectors
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US6995006B2 (en) 1997-09-05 2006-02-07 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
PT1944362E (en) 1997-09-05 2016-01-27 Genzyme Corp Methods for generating high titer helper-free preparations of recombinant aav vectors
CA2303768C (en) 1997-09-19 2009-11-24 The Trustees Of The University Of Pennsylvania Methods and vector constructs useful for production of recombinant aav
EP1015619A1 (en) * 1997-09-19 2000-07-05 The Trustees Of The University Of Pennsylvania Methods and cell line useful for production of recombinant adeno-associated viruses
US6346415B1 (en) 1997-10-21 2002-02-12 Targeted Genetics Corporation Transcriptionally-activated AAV inverted terminal repeats (ITRS) for use with recombinant AAV vectors
US6642051B1 (en) 1997-10-21 2003-11-04 Targeted Genetics Corporation Amplifiable adeno-associated virus(AAV) packaging cassettes for the production of recombinant AAV vectors
IT1297074B1 (en) 1997-11-21 1999-08-03 Angeletti P Ist Richerche Bio HORMONE-DEPENDENT FORMS OF THE REP PROTEIN OF THE ADENUS ASSOCIATED VIRUS (AAV-2), DNA SEQUENCES CODING FOR THEM, VECTORS THAT
GB9811172D0 (en) * 1998-05-22 1998-07-22 Royal Free Hosp School Med Virus production
DK1930418T3 (en) 1998-09-04 2015-07-13 Genzyme Corp Methods for Generating Helper-Free High Titer Preparations of Released Recombinant AAV Vectors
DE19905501B4 (en) 1999-02-10 2005-05-19 MediGene AG, Gesellschaft für molekularbiologische Kardiologie und Onkologie A method of producing a recombinant adeno-associated virus, suitable compositions therefor, and use for the manufacture of a medicament
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
EP1939300A1 (en) 1999-05-28 2008-07-02 Targeted Genetics Corporation Methods and compositions for lowering the level of tumor necrosis factor (TNF) in TNF-associated disorders
US6537540B1 (en) 1999-05-28 2003-03-25 Targeted Genetics Corporation Methods and composition for lowering the level of tumor necrosis factor (TNF) in TNF-associated disorders
AU781958C (en) 1999-08-09 2006-03-30 Targeted Genetics Corporation Enhancement of expression of a single-stranded, heterologous nucleotide sequence from recombinant viral vectors by designing the sequence such that it forms intrastrand base pairs
EP1916258B1 (en) 1999-08-09 2014-04-23 Targeted Genetics Corporation Enhancement of expression of a single-stranded, heterologous nucleotide sequence from recombinant viral vectors by designing the sequence such that it forms intrastrand base pairs
US7115391B1 (en) 1999-10-01 2006-10-03 Genovo, Inc. Production of recombinant AAV using adenovirus comprising AAV rep/cap genes
US7271002B2 (en) 2001-11-09 2007-09-18 United States Of America, Represented By The Secretary, Department Of Health And Human Services Production of adeno-associated virus in insect cells
US6723551B2 (en) 2001-11-09 2004-04-20 The United States Of America As Represented By The Department Of Health And Human Services Production of adeno-associated virus in insect cells
EP1587923B1 (en) 2003-01-22 2011-08-24 Duke University Improved constructs for expressing lysosomal polypeptides
CA2555675A1 (en) 2004-02-10 2005-08-25 Trustees Of Dartmouth College Nicotinamide riboside kinase compositions and methods for using the same
AU2006304997B2 (en) 2005-10-20 2012-03-01 Uniqure Ip B.V. Improved AAV vectors produced in insect cells
ES2785223T3 (en) 2006-06-21 2020-10-06 Uniqure Ip Bv Insect cells for AAV vector production
WO2008024998A2 (en) 2006-08-24 2008-02-28 Virovek, Inc. Expression in insect cells of genes with overlapping open reading frames, methods and compositions therefor
WO2008067480A2 (en) 2006-11-29 2008-06-05 Nationwide Children's Hospital Myostatin inhibition for enhancing muscle and/or improving muscle function
US11219696B2 (en) 2008-12-19 2022-01-11 Nationwide Children's Hospital Delivery of polynucleotides using recombinant AAV9
US9415121B2 (en) 2008-12-19 2016-08-16 Nationwide Children's Hospital Delivery of MECP2 polynucleotide using recombinant AAV9
EP2412387B1 (en) 2009-03-27 2014-11-19 Proyecto de Biomedicina Cima, S.L. Methods and compositions for the treatment of cirrhosis and liver fibrosis
WO2010129021A1 (en) 2009-05-02 2010-11-11 Genzyme Corporation Gene therapy for neurodegenerative disorders
BR112012010755A2 (en) 2009-11-05 2015-09-22 Proyecto Biomedicina Cima Sl gene construct allowing inducible hepato-specific expression of a polynucleotide of interest in response to an inducing agent, vector, recombinant viral genome, virion, in vitro method for expression of a polynucleotide of interest in a cell of hepatic origin, composition pharmaceutical and inducible bi-directional promoter-operator suitable for the inducible hepato-specific expression of two polynucleotides of interest by an inducing agent
CN108744262A (en) 2010-11-23 2018-11-06 普莱萨格生命科学公司 Treatment and composition for for physical delivery
US20140155469A1 (en) 2011-04-19 2014-06-05 The Research Foundation Of State University Of New York Adeno-associated-virus rep sequences, vectors and viruses
US10196636B2 (en) 2011-04-21 2019-02-05 Nationwide Children's Hospital, Inc. Recombinant virus products and methods for inhibition of expression of myotilin
EP2699673B1 (en) 2011-04-21 2018-09-19 Nationwide Children's Hospital, Inc. Recombinant virus products and methods for inhibition of expression of myotilin
US20130039888A1 (en) 2011-06-08 2013-02-14 Nationwide Children's Hospital Inc. Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
AU2012286994B2 (en) 2011-07-25 2017-08-10 Nationwide Children's Hospital, Inc. Recombinant virus products and methods for inhibition of expression of DUX4
US9434928B2 (en) 2011-11-23 2016-09-06 Nationwide Children's Hospital, Inc. Recombinant adeno-associated virus delivery of alpha-sarcoglycan polynucleotides
US9163259B2 (en) 2012-05-04 2015-10-20 Novartis Ag Viral vectors for the treatment of retinal dystrophy
EP3769789A1 (en) 2012-08-01 2021-01-27 Nationwide Children's Hospital Intrathecal delivery of recombinant adeno-associated virus 9
JP2015529685A (en) 2012-09-17 2015-10-08 ザ・リサーチ・インスティテュート・アット・ネイションワイド・チルドレンズ・ホスピタルThe Research Institute Atnationwide Children’S Hospital Compositions and methods for the treatment of amyotrophic lateral sclerosis
JP6576904B2 (en) 2013-04-04 2019-09-18 トラスティーズ・オブ・ダートマス・カレッジ Compositions and methods for in vivo excision of HIV-1 proviral DNA
EP2983707B1 (en) 2013-04-08 2019-06-12 University of Iowa Research Foundation Chimeric adeno-associated virus/ bocavirus parvovirus vector
KR102268473B1 (en) 2013-04-20 2021-06-25 더 리서치 인스티튜트 앳 네이션와이드 칠드런스 하스피탈 Recombinant adeno-associated virus delivery of exon 2-targeted us7nrna polynucleotide constructs
CN115120746A (en) 2013-05-15 2022-09-30 明尼苏达大学董事会 Adeno-associated virus-mediated gene transfer to the central nervous system
HRP20230015T1 (en) 2013-08-27 2023-02-17 Research Institute At Nationwide Children's Hospital Products and methods for treatment of amyotrophic lateral sclerosis
WO2015069647A1 (en) 2013-11-05 2015-05-14 The Research Institute At Nationwide Children's Hospital COMPOSITIONS AND METHODS FOR INHIBITING NF- kB AND SOD-1 TO TREAT AMYOTROPHIC LATERAL SCLEROSIS
US10047130B2 (en) 2014-03-18 2018-08-14 Washington University Methods and compositions for red-shifted chromophore substitution for optogenetic applications
WO2015191508A1 (en) 2014-06-09 2015-12-17 Voyager Therapeutics, Inc. Chimeric capsids
US11053494B2 (en) 2014-08-09 2021-07-06 Research Institute At Nationwide Children's Hospital Methods and materials for activating an internal ribosome entry site in exon 5 of the DMD gene
WO2016057975A2 (en) 2014-10-10 2016-04-14 Research Institute At Nationwide Children's Hospital Guided injections for aav gene transfer to muscle
EP3215602B1 (en) 2014-11-05 2019-12-25 The Research Institute at Nationwide Children's Hospital Methods and materials for producing recombinant viruses in eukaryotic microalgae
WO2016094783A1 (en) 2014-12-12 2016-06-16 Voyager Therapeutics, Inc. Compositions and methods for the production of scaav
CN107427666B (en) 2015-01-30 2022-11-04 加利福尼亚大学校董 Subpial gene delivery system for spinal cord
MA41451A (en) 2015-02-04 2017-12-12 Univ Washington ANTI-TAU CONSTRUCTIONS
EP3262172A2 (en) 2015-02-23 2018-01-03 Crispr Therapeutics AG Materials and methods for treatment of hemoglobinopathies
KR20220082100A (en) 2015-05-15 2022-06-16 리젠츠 오브 더 유니버시티 오브 미네소타 Adeno-associated for therapeutic delivery to central nervous system
US10017832B2 (en) 2015-08-25 2018-07-10 Washington University Compositions and methods for site specific recombination at asymmetric sites
AU2016323575B2 (en) 2015-09-17 2022-10-27 Research Institute At Nationwide Children's Hospital Methods and materials for GALGT2 gene therapy
CA2998287A1 (en) 2015-09-24 2017-04-20 Crispr Therapeutics Ag Novel family of rna-programmable endonucleases and their uses in genome editing and other applications
US20180230489A1 (en) 2015-10-28 2018-08-16 Voyager Therapeutics, Inc. Regulatable expression using adeno-associated virus (aav)
US11369692B2 (en) 2015-10-28 2022-06-28 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of Duchenne Muscular Dystrophy
EP3371305A1 (en) 2015-11-06 2018-09-12 Crispr Therapeutics AG Materials and methods for treatment of glycogen storage disease type 1a
BR112018009954A2 (en) 2015-11-16 2018-11-13 Res Inst Nationwide Childrens Hospital materials and methods for the treatment of titin-based myopathies and other titinopathies
EP3384024B1 (en) 2015-12-01 2022-02-02 CRISPR Therapeutics AG Materials and methods for treatment of alpha-1 antitrypsin deficiency
BR112018012894A2 (en) 2015-12-23 2018-12-04 Crispr Therapeutics Ag Materials and Methods for Treatment of Amyotrophic Lateral Sclerosis and / or Frontotemporal Lobular Degeneration
US11938193B2 (en) 2016-01-08 2024-03-26 Washington University Compositions comprising chemerin and methods of use thereof
EP3411078A1 (en) 2016-02-02 2018-12-12 Crispr Therapeutics AG Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
WO2017139381A1 (en) 2016-02-08 2017-08-17 University Of Iowa Research Foundation Methods to produce chimeric adeno-associated virus/bocavirus parvovirus
WO2017141109A1 (en) 2016-02-18 2017-08-24 Crispr Therapeutics Ag Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
WO2017147509A1 (en) 2016-02-25 2017-08-31 Marco Colonna Compositions comprising trem2 and methods of use thereof
CA3015633A1 (en) 2016-02-26 2017-08-31 Research Institute At Nationwide Children's Hospital Recombinant virus products and methods for inducing dux4 exon skipping
WO2017158422A1 (en) 2016-03-16 2017-09-21 Crispr Therapeutics Ag Materials and methods for treatment of hereditary haemochromatosis
PT3436593T (en) 2016-03-28 2023-01-31 Ultragenyx Pharmaceutical Inc Methods of heat inactivation of adenovirus
EP3436427B1 (en) 2016-03-28 2021-12-08 The Regents of the University of California Method and composition for treating neuronal hyper-excitability
WO2017173411A1 (en) 2016-04-02 2017-10-05 Research Institute At Nation Wide Children's Hospital Modified u6 promoter system for tissue specific expression
EP3442600B1 (en) 2016-04-15 2024-03-13 Research Institute at Nationwide Children's Hospital Adeno-associated virus vector delivery of b-sarcoglycan and microrna-29 and the treatment of muscular dystrophy
MA45477A (en) 2016-04-15 2019-02-20 Res Inst Nationwide Childrens Hospital ADENOASSOCIATED VIRUS VECTOR VECTOR MICROARN-29 AND MICRO-DYSTROPHINE TO TREAT MUSCLE DYSTROPHY
CN109715198B (en) 2016-04-18 2022-09-06 克里斯珀医疗股份公司 Materials and methods for treating hemoglobinopathies
EP3448987A4 (en) 2016-04-29 2020-05-27 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
WO2017191503A1 (en) 2016-05-05 2017-11-09 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
RU2764587C2 (en) 2016-05-18 2022-01-18 Вояджер Терапьютикс, Инк. Methods and compositions for treatment of huntington's chorea
SG11201809699XA (en) 2016-05-18 2018-12-28 Voyager Therapeutics Inc Modulatory polynucleotides
US11427838B2 (en) 2016-06-29 2022-08-30 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of myotonic dystrophy type 1 (DM1) and other related disorders
EP3478313B1 (en) 2016-06-29 2022-05-04 CRISPR Therapeutics AG Materials and methods for treatment of amyotrophic lateral sclerosis (als) and other related disorders
CA3029119A1 (en) 2016-06-29 2018-01-04 Crispr Therapeutics Ag Materials and methods for treatment of friedreich ataxia and other related disorders
CA3029132A1 (en) 2016-07-06 2018-01-11 Crispr Therapeutics Ag Materials and methods for treatment of pain related disorders
EP3481857A1 (en) 2016-07-06 2019-05-15 Crispr Therapeutics AG Materials and methods for treatment of pain related disorders
WO2018007871A1 (en) 2016-07-08 2018-01-11 Crispr Therapeutics Ag Materials and methods for treatment of transthyretin amyloidosis
WO2018020323A2 (en) 2016-07-25 2018-02-01 Crispr Therapeutics Ag Materials and methods for treatment of fatty acid disorders
US11795473B2 (en) 2016-10-14 2023-10-24 Ultragenyx Pharmaceutical Inc. Use of tonicifying agents to enhance recombinant adeno-associated virus yield
EP3541946A1 (en) 2016-11-15 2019-09-25 Regents Of The University Of Minnesota Method for improving neurological function in mpsi and mpsii and other neurological disorders
MX2019005874A (en) 2016-11-17 2020-02-07 Nationwide Childrens Hospital Inc Intrathecal delivery of recombinant adeno-associated virus encoding methyl-cpg binding protein 2.
CA3045667A1 (en) 2016-12-13 2018-06-21 Seattle Children's Hospital (dba Seattle Children's Research Institute) Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo
WO2018132747A1 (en) 2017-01-13 2018-07-19 University Of Iowa Research Foundation Bocaparvovirus small noncoding rna and uses thereof
US20200040061A1 (en) 2017-02-22 2020-02-06 Crispr Therapeutics Ag Materials and methods for treatment of early onset parkinson's disease (park1) and other synuclein, alpha (snca) gene related conditions or disorders
EP3585895A1 (en) 2017-02-22 2020-01-01 CRISPR Therapeutics AG Compositions and methods for gene editing
US11407997B2 (en) 2017-02-22 2022-08-09 Crispr Therapeutics Ag Materials and methods for treatment of primary hyperoxaluria type 1 (PH1) and other alanine-glyoxylate aminotransferase (AGXT) gene related conditions or disorders
US11559588B2 (en) 2017-02-22 2023-01-24 Crispr Therapeutics Ag Materials and methods for treatment of Spinocerebellar Ataxia Type 1 (SCA1) and other Spinocerebellar Ataxia Type 1 Protein (ATXN1) gene related conditions or disorders
US20200216857A1 (en) 2017-02-22 2020-07-09 Crispr Therapeutics Ag Materials and methods for treatment of spinocerebellar ataxia type 2 (sca2) and other spinocerebellar ataxia type 2 protein (atxn2) gene related conditions or disorders
SG11201908575SA (en) 2017-03-17 2019-10-30 Res Inst Nationwide Childrens Hospital Adeno-associated virus vector delivery of muscle specific micro-dystrophin to treat muscular dystrophy
EP3596112A2 (en) 2017-03-17 2020-01-22 Newcastle University Adeno-associated virus vector delivery of a fragment of micro-dystrophin to treat muscular dystrophy
US11898170B2 (en) 2017-03-22 2024-02-13 Ultragenyx Pharmaceutical Inc. Cell culture methods involving HDAC inhibitors or rep proteins
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
SG11201909868YA (en) 2017-05-05 2019-11-28 Voyager Therapeutics Inc Compositions and methods of treating huntington's disease
EP3621981A2 (en) 2017-05-12 2020-03-18 CRISPR Therapeutics AG Materials and methods for engineering cells and uses thereof in immuno-oncology
EP3652324A1 (en) 2017-07-08 2020-05-20 Genethon Treatment of spinal muscular atrophy
CN111132626B (en) 2017-07-17 2024-01-30 沃雅戈治疗公司 Track array guidance system
KR20200044793A (en) 2017-08-03 2020-04-29 보이저 테라퓨틱스, 인크. Compositions and methods for delivery of AAV
US20200231986A1 (en) 2017-09-29 2020-07-23 Massachusetts Eye And Ear Infirmary Production of adeno-associated viruses in insect cells
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)
AU2018353012A1 (en) 2017-10-17 2020-04-23 Bayer Healthcare Llc Compositions and methods for gene editing for Hemophilia A
WO2019078916A1 (en) 2017-10-18 2019-04-25 Research Institute At Nationwide Children's Hospital Adeno-associated virus vector delivery of muscle specific micro-dystrophin to treat muscular dystrophy
EP3697916A4 (en) 2017-10-20 2021-08-11 Research Institute at Nationwide Children's Hospital Methods and materials for nt-3 gene therapy
MA50849A (en) 2017-10-26 2020-09-02 Vertex Pharma SUBSTANCES AND METHODS FOR THE TREATMENT OF HEMOGLOBINOPATHIES
KR20200086292A (en) 2017-11-08 2020-07-16 아벡시스, 인크. Means and methods for producing viral vectors and uses thereof
MA50578A (en) 2017-11-09 2021-09-15 Vertex Pharma CRISPR / CAS SYSTEMS FOR THE TREATMENT OF DMD
MA50877A (en) 2017-11-21 2020-09-30 Bayer Healthcare Llc MATERIALS AND METHODS FOR THE TREATMENT OF AUTOSOMIC DOMINANT PIGMENTAL RETINITIS
WO2019118935A1 (en) 2017-12-14 2019-06-20 Casebia Therapeutics Limited Liability Partnership Novel rna-programmable endonuclease systems and their use in genome editing and other applications
EP3728595A1 (en) 2017-12-21 2020-10-28 CRISPR Therapeutics AG Materials and methods for treatment of usher syndrome type 2a and/or non-syndromic autosomal recessive retinitis pigmentosa (arrp)
JP7402163B2 (en) 2017-12-21 2023-12-20 クリスパー セラピューティクス アーゲー Materials and methods for the treatment of Usher syndrome type 2A
US20210254057A1 (en) 2018-01-12 2021-08-19 Crispr Therapeutics Ag Compositions and methods for gene editing by targeting transferrin
US11566236B2 (en) 2018-02-05 2023-01-31 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
EP3749768A1 (en) 2018-02-05 2020-12-16 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
WO2019161310A1 (en) 2018-02-16 2019-08-22 Casebia Therapeutics Limited Liability Partnership Compositions and methods for gene editing by targeting fibrinogen-alpha
WO2019183150A1 (en) 2018-03-19 2019-09-26 Casebia Therapeutics Limited Liability Partnership Novel rna-programmable endonuclease systems and uses thereof
WO2019204668A1 (en) 2018-04-18 2019-10-24 Casebia Therapeutics Limited Liability Partnership Compositions and methods for knockdown of apo(a) by gene editing for treatment of cardiovascular disease
AU2019257755A1 (en) 2018-04-27 2020-11-12 Voyager Therapeutics, Inc. Methods for measuring the potency of AADC viral vectors
US20210230632A1 (en) 2018-05-15 2021-07-29 Voyager Therapeutics, Inc. Compositions and methods for delivery of aav
SG11202011296VA (en) 2018-05-15 2020-12-30 Voyager Therapeutics Inc Compositions and methods for the treatment of parkinson's disease
US20210214749A1 (en) 2018-05-16 2021-07-15 Voyager Therapeutics, Inc. Directed evolution
WO2019222441A1 (en) 2018-05-16 2019-11-21 Voyager Therapeutics, Inc. Aav serotypes for brain specific payload delivery
EP3801638A1 (en) 2018-06-08 2021-04-14 Novartis AG Cell-based assay for measuring drug product potency
GB201809588D0 (en) 2018-06-12 2018-07-25 Univ Bristol Materials and methods for modulating intraocular and intracranial pressure
EP3807404A1 (en) 2018-06-13 2021-04-21 Voyager Therapeutics, Inc. Engineered 5' untranslated regions (5' utr) for aav production
CA3104471A1 (en) 2018-06-18 2019-12-26 Research Institute At Nationwide Children's Hospital Recombinant adeno-associated virus products and methods for treating dystroglycanopathies and laminin-deficient muscular dystrophies
BR112020025995A2 (en) 2018-06-18 2021-03-23 Research Institute At Nationwide Children's Hospital administration of muscle-specific microdystrophin by adeno-associated virus vector to treat muscular dystrophy
MX2020014119A (en) 2018-06-29 2021-06-18 Res Inst Nationwide Childrens Hospital Recombinant adeno-associated virus products and methods for treating limb girdle muscular dystrophy 2a.
CN112567035A (en) 2018-07-02 2021-03-26 沃雅戈治疗公司 Treatment of amyotrophic lateral sclerosis and spinal cord related disorders
US20210355454A1 (en) 2018-07-24 2021-11-18 Voyager Therapeutics, Inc. Systems and methods for producing gene therapy formulations
US20210324417A1 (en) 2018-08-29 2021-10-21 Research Institute At Nationwide Children's Hospital Products and methods for inhibition of expression of mutant gars protein
KR20210068068A (en) 2018-09-28 2021-06-08 보이저 테라퓨틱스, 인크. Prataxin expression constructs with engineered promoters and methods of use thereof
US20210348242A1 (en) 2018-10-04 2021-11-11 Voyager Therapeutics, Inc. Methods for measuring the titer and potency of viral vector particles
TW202028458A (en) 2018-10-05 2020-08-01 美商航海家醫療公司 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
EP3867389A1 (en) 2018-10-15 2021-08-25 Voyager Therapeutics, Inc. Expression vectors for large-scale production of raav in the baculovirus/sf9 system
MX2021004455A (en) 2018-10-17 2021-08-11 Crispr Therapeutics Ag Compositions and methods for delivering transgenes.
BR112021009739A2 (en) 2018-11-30 2021-10-19 Novartis Ag AAV VIRAL VECTORS AND USES THEREOF
US20220042045A1 (en) 2018-12-21 2022-02-10 Genethon Expression cassettes for gene therapy vectors
KR20210110345A (en) 2018-12-31 2021-09-07 더 리서치 인스티튜트 앳 네이션와이드 칠드런스 하스피탈 DUX4 RNA Silencing Using RNA Targeting CRISPR-Cas13b
AU2020208467A1 (en) 2019-01-18 2021-08-05 Voyager Therapeutics, Inc. Methods and systems for producing AAV particles
KR20210130158A (en) * 2019-01-31 2021-10-29 오레곤 헬스 앤드 사이언스 유니버시티 Methods Using Transcription-Dependent Directed Evolution of AAV Capsids
TW202045530A (en) 2019-02-04 2020-12-16 美國全美兒童醫院之研究學會 Adeno-associated virus delivery of cln3 polynucleotide
CA3127801A1 (en) 2019-02-04 2020-08-13 Research Institute At Nationwide Children's Hospital Adeno-associated virus delivery of cln6 polynucleotide
SG11202108357PA (en) 2019-02-15 2021-08-30 Crispr Therapeutics Ag Gene editing for hemophilia a with improved factor viii expression
CR20210444A (en) 2019-02-25 2021-11-02 Novartis Ag Compositions and methods to treat bietti crystalline dystrophy
WO2020174369A2 (en) 2019-02-25 2020-09-03 Novartis Ag Compositions and methods to treat bietti crystalline dystrophy
WO2020176614A1 (en) 2019-02-26 2020-09-03 Research Institute At Nationwide Children's Hospital Adeno-associated virus vector delivery of b-sarcoglycan and the treatment of muscular dystrophy
EP3937963A2 (en) 2019-03-12 2022-01-19 CRISPR Therapeutics AG Novel high fidelity rna-programmable endonuclease systems and uses thereof
SG11202110904PA (en) 2019-04-12 2021-10-28 Ultragenyx Pharmaceutical Inc Engineered producer cell lines and methods of making and using the same
AU2020257182A1 (en) 2019-04-15 2021-12-09 Spirovant Sciences, Inc. Methods and compositions for transgene expression
WO2020214737A1 (en) 2019-04-15 2020-10-22 Sanford Research Gene therapy for treating or preventing visual effects in batten disease
TW202106879A (en) 2019-04-29 2021-02-16 美商航海家醫療公司 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
WO2020225606A1 (en) 2019-05-08 2020-11-12 Crispr Therapeutics Ag Crispr/cas all-in-two vector systems for treatment of dmd
US20220226507A1 (en) 2019-05-17 2022-07-21 Research Institute At Nationwide Children's Hospital Optimized gene therapy targeting retinal cells
WO2020264254A1 (en) 2019-06-28 2020-12-30 Crispr Therapeutics Ag Materials and methods for controlling gene editing
JP2022541070A (en) 2019-07-25 2022-09-21 ノバルティス アーゲー Regulatable expression system
EP4010465A1 (en) 2019-08-09 2022-06-15 Voyager Therapeutics, Inc. Cell culture medium for use in producing gene therapy products in bioreactors
EP4017871B1 (en) 2019-08-21 2024-01-17 Research Institute at Nationwide Children's Hospital Adeno-associated virus vector delivery of alpha-sarcoglycan and the treatment of muscular dystrophy
US20220364114A1 (en) 2019-08-26 2022-11-17 Voyager Therapeutics, Inc. 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
WO2021072129A2 (en) 2019-10-08 2021-04-15 Trustees Of Boston College Proteins containing multiple, different unnatural amino acids and methods of making and using such proteins
WO2021072115A1 (en) 2019-10-08 2021-04-15 Regents Of The University Of Minnesota Crispr-mediated human genome editing with vectors
AU2020366242A1 (en) 2019-10-18 2022-05-26 Research Institute At Nationwide Children's Hospital Materials and methods for the treatment of disorders associated mutations in the IRF2BPL gene
JP2022552015A (en) 2019-10-18 2022-12-14 リサーチ インスティチュート アット ネイションワイド チルドレンズ ホスピタル Gene therapy targeting cochlear cells
IL293210A (en) 2019-11-22 2022-07-01 Res Inst Nationwide Childrens Hospital Materials and methods for treatment of disorders associated with the ighmbp2 gene
CN115516100A (en) 2019-12-20 2022-12-23 全国儿童医院研究所 Optimized gene therapy targeting muscles in muscle diseases
CA3171959A1 (en) 2020-02-18 2021-08-26 Research Institute At Nationwide Children's Hospital Aav-mediated targeting of mirna in the treatment of x-linked disorders
KR20220150353A (en) 2020-03-05 2022-11-10 네오티엑스 테라퓨틱스 엘티디. Methods and compositions for treating cancer using immune cells
BR112022018658A2 (en) 2020-03-16 2022-12-20 Ultragenyx Pharmaceutical Inc METHODS TO ENHANCE THE YIELD OF RECOMBINANT ADENO-ASSOCIATED VIRUSES
CN115379863A (en) 2020-04-14 2022-11-22 吉尼松公司 Carrier for treating acid ceramidase deficiency
BR112022020753A2 (en) 2020-04-15 2022-12-20 Voyager Therapeutics Inc TAU-BINDING COMPOUNDS
WO2021247995A2 (en) 2020-06-04 2021-12-09 Voyager Therapeutics, Inc. Compositions and methods of treating neuropathic pain
CN116348149A (en) 2020-06-15 2023-06-27 全国儿童医院研究所 Adeno-associated viral vector delivery for muscular dystrophy
US20230374483A1 (en) 2020-07-08 2023-11-23 Regents Of The University Of Minnesota Modified hexosaminidase and uses thereof
WO2022018638A1 (en) 2020-07-21 2022-01-27 Crispr Therapeutics Ag Genome-editing compositions and methods to modulate faah for treatment of neurological disorders
US20230227802A1 (en) 2020-07-27 2023-07-20 Voyager Therapeutics, Inc. Compositions and methods for the treatment of neurological disorders related to glucosylceramidase beta deficiency
WO2022026410A2 (en) 2020-07-27 2022-02-03 Voyager Therapeutics, Inc Compositions and methods for the treatment of niemann-pick type c1 disease
EP4192514A1 (en) 2020-08-06 2023-06-14 Voyager Therapeutics, Inc. Cell culture medium for use in producing gene therapy products in bioreactors
WO2022047201A1 (en) 2020-08-27 2022-03-03 University Of Iowa Research Foundation Gene knock-out for treatment of glaucoma
TW202227634A (en) 2020-09-08 2022-07-16 美商薩羅塔治療公司 Systemic delivery of adeno-associated virus vector expressing g-sarcoglycan and the treatment of muscular dystrophy
EP4214317A2 (en) 2020-09-15 2023-07-26 Research Institute at Nationwide Children's Hospital Aav-mediated homology-independent targeted integration gene editing for correction of diverse dmd mutations in patients with muscular dystrophy
JP2023543029A (en) 2020-09-28 2023-10-12 リサーチ インスティチュート アット ネイションワイド チルドレンズ ホスピタル Products and methods for the treatment of muscular dystrophy
WO2022070107A1 (en) 2020-09-30 2022-04-07 Crispr Therapeutics Ag Materials and methods for treatment of amyotrophic lateral sclerosis
AU2021372262A1 (en) 2020-11-02 2023-06-01 Biomarin Pharmaceutical Inc. Process for enriching adeno-associated virus
WO2022115745A1 (en) 2020-11-30 2022-06-02 Research Institute At Nationwide Children's Hospital Compositions and methods for treating facioscapulohumeral muscular dystrophy (fshd)
WO2022133246A1 (en) 2020-12-17 2022-06-23 Vertex Pharmaceuticals Incorporated Compositions and methods for editing beta-globin for treatment of hemaglobinopathies
CA3209471A1 (en) 2021-01-27 2022-08-04 Research Institute At Nationwide Children's Hospital Materials and methods for the treatment of lysosomal acid lipase deficiency (lal-d)
CA3210662A1 (en) 2021-02-03 2022-08-11 Research Institute At Nationwide Children's Hospital Compositions and methods for treating disease associated with dux4 overexpression
WO2022170082A1 (en) 2021-02-05 2022-08-11 Regents Of The University Of Minnesota Methods for preventing cardiac or skeletal defects in diseases including mucopolysaccharidoses
WO2022170038A1 (en) 2021-02-05 2022-08-11 Amicus Therapeutics, Inc. Adeno-associated virus delivery of cln3 polynucleotide
EP4301462A1 (en) 2021-03-04 2024-01-10 Research Institute at Nationwide Children's Hospital Products and methods for treatment of dystrophin-based myopathies using crispr-cas9 to correct dmd exon duplications
CN115427561A (en) 2021-03-09 2022-12-02 辉大(上海)生物科技有限公司 Engineered CRISPR/Cas13 systems and uses thereof
WO2022221424A1 (en) 2021-04-13 2022-10-20 Research Institute At Nationwide Children's Hospital Recombinant adeno-associated virus encoding methyl-cpg binding protein 2 for treating pitt hopkins syndrome via intrathecal delivery
WO2022226334A1 (en) 2021-04-23 2022-10-27 Research Institute At Nationwide Children's Hospital Products and methods for treating muscular dystrophy
CA3218195A1 (en) 2021-05-07 2022-11-10 Robin Ali Abca4 genome editing
CA3218350A1 (en) 2021-05-17 2022-11-24 Sarepta Therapeutics, Inc. Production of recombinant aav vectors for treating muscular dystrophy
EP4108263A3 (en) 2021-06-02 2023-03-22 Research Institute at Nationwide Children's Hospital Recombinant adeno-associated virus products and methods for treating limb girdle muscular dystrophy 2a
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
BR112023023768A2 (en) 2021-06-11 2024-02-27 Bayer Ag TYPE V RNA PROGRAMMABLE ENDONUCLEASE SYSTEMS
WO2023283962A1 (en) 2021-07-16 2023-01-19 Huigene Therapeutics Co., Ltd. Modified aav capsid for gene therapy and methods thereof
CA3228365A1 (en) 2021-08-11 2023-02-16 Solid Biosciences Inc. Treatment of muscular dystrophy
WO2023034989A1 (en) 2021-09-03 2023-03-09 Biomarin Pharmaceutical Inc. Aav capsid compositions and methods for delivery
WO2023034996A1 (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
WO2023034980A1 (en) 2021-09-03 2023-03-09 Bomarin 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
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
WO2023042104A1 (en) 2021-09-16 2023-03-23 Novartis Ag Novel transcription factors
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
AU2022360382A1 (en) 2021-10-07 2024-05-09 Research Institute At Nationwide Children's Hospital Products and methods for myelin protein zero silencing and treating cmt1b disease
WO2023060233A1 (en) 2021-10-08 2023-04-13 Amicus Therapeutics, Inc. Biomarkers for lysosomal storage diseases
JP2023059858A (en) 2021-10-15 2023-04-27 リサーチ インスティチュート アット ネイションワイド チルドレンズ ホスピタル Self-complementary adeno-associated virus vector and use thereof in treatment of muscular dystrophy
WO2023077078A1 (en) 2021-10-29 2023-05-04 Ultragenyx Pharmaceutical Inc. Engineered cell lines for increased production of recombinant adeno-associated virus (raav)
WO2023091949A2 (en) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions and methods for the treatment of neurological disorders related to glucosylceramidase beta deficiency
JP2023081369A (en) 2021-11-30 2023-06-09 リサーチ インスティチュート アット ネイションワイド チルドレンズ ホスピタル Self-complementary adeno-associated virus vector and its use in treatment of muscular dystrophy
EP4198048A1 (en) 2021-12-16 2023-06-21 Genethon Calpain-3 gene transfer increase using modified itr sequences
EP4198047A1 (en) 2021-12-16 2023-06-21 Genethon Fukutin related protein gene transfer increase using modified itr sequences
EP4198046A1 (en) 2021-12-16 2023-06-21 Genethon Alpha-sarcoglycan gene transfer increase using modified itr sequences
EP4198134A1 (en) 2021-12-16 2023-06-21 Genethon Gamma-sarcoglycan gene transfer increase using modified itr sequences
WO2023122669A1 (en) 2021-12-21 2023-06-29 Research Institute At Nationwide Children's Hospital Materials and methods for the treatment of limb girdle muscular dystrophy
WO2023118068A1 (en) 2021-12-23 2023-06-29 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2023168400A2 (en) 2022-03-03 2023-09-07 Research Institute At Nationwide Children's Hospital Materials and methods for the treatment of eif2b5 mutations and diseases resulting therefrom
WO2023178067A1 (en) * 2022-03-16 2023-09-21 Rutgers, The State University Of New Jersey Controlled muscle‐specific gene delivery
WO2023196818A1 (en) 2022-04-04 2023-10-12 The Regents Of The University Of California Genetic complementation compositions and methods
WO2023214346A1 (en) 2022-05-06 2023-11-09 Novartis Ag Novel recombinant aav vp2 fusion polypeptides
WO2023240177A1 (en) 2022-06-08 2023-12-14 Research Instiitute At Nationwide Children's Hospital Products and methods for treating diseases or conditions associated with mutant or pathogenic kcnq3 expression
WO2023240236A1 (en) 2022-06-10 2023-12-14 Voyager Therapeutics, Inc. Compositions and methods for the treatment of spinal muscular atrophy related disorders
WO2023237587A1 (en) 2022-06-10 2023-12-14 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2024011115A1 (en) 2022-07-06 2024-01-11 Research Institute At Nationwide Children's Hospital Adeno-associated virus delivery of cln1 polynucleotide
WO2024035782A1 (en) 2022-08-10 2024-02-15 Aav Gene Therapeutics, Inc. Aav-mediated intramuscular delivery of insulin
WO2024059739A1 (en) 2022-09-15 2024-03-21 Voyager Therapeutics, Inc. Tau binding compounds
WO2024064913A1 (en) 2022-09-23 2024-03-28 Sarepta Therapeutics, Inc. Recombinant aav vectors for treating muscular dystrophy
WO2024081706A1 (en) 2022-10-11 2024-04-18 Research Institute At Nationwide Children's Hospital Adeno-associated virus delivery to treat spinal muscular atrophy with respiratory distress type 1 (smard1) and charcot-marie-tooth type 2s (cmt2s)
WO2024092171A1 (en) 2022-10-26 2024-05-02 University Of Iowa Research Foundation Method to deliver large genes using virus and a dna recombination system
WO2024092126A1 (en) 2022-10-27 2024-05-02 Cargo Therapeutics, Inc. Compositions and methods for improved immunotherapies

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5354678A (en) * 1990-10-30 1994-10-11 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5478745A (en) * 1992-12-04 1995-12-26 University Of Pittsburgh Recombinant viral vector system
EP0728214B1 (en) * 1993-11-09 2004-07-28 Medical College Of Ohio Stable cell lines capable of expressing the adeno-associated virus replication gene
DE69433592T2 (en) * 1993-11-09 2005-02-10 Targeted Genetics Corp., Seattle THE ACHIEVEMENT OF HIGH TITERS OF THE RECOMBINANT AAV VECTOR

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5354678A (en) * 1990-10-30 1994-10-11 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors

Also Published As

Publication number Publication date
EP0796339A1 (en) 1997-09-24
AU4596396A (en) 1996-06-26
JPH10511264A (en) 1998-11-04
WO1996017947A1 (en) 1996-06-13
CA2207927A1 (en) 1996-06-13

Similar Documents

Publication Publication Date Title
AU707866B2 (en) Packaging cell lines for generation of high titers of recombinant AAV vectors
US6924128B2 (en) Packaging cell lines for generation of high titers of recombinant AAV vectors
WO1996017947A9 (en) Packaging cell lines for generation of high titers of recombinant aav vectors
US6541258B2 (en) AAV split-packaging genes and cell lines comprising such genes for use in the production of recombinant AAV vectors
AU688428B2 (en) Generation of high titers of recombinant AAV vectors
US6936466B2 (en) Transcriptionally-activated AAV inverted terminal repeats (ITRs) for use with recombinant AAV vectors
US6642051B1 (en) Amplifiable adeno-associated virus(AAV) packaging cassettes for the production of recombinant AAV vectors
WO1998027204A9 (en) Aav split-packaging genes and cell lines comprising such genes for use in the production of recombinant aav vectors
CA2269661A1 (en) Recombinase-activatable aav packaging cassettes for use in the production of aav vectors
CA2304801C (en) Transcriptionally-activated aav inverted terminal repeats (itrs) for use with recombinant aav vectors
AU758541B2 (en) Amplifiable adeno-associated virus (AAV) packaging cassettes for the production of recombinant AAV vectors
AU2003203790B2 (en) Transcriptionally-activated AAV inverted terminal repeats (ITRs) for use with recombinant AAV vectors