CA2098483C - Safe adeno-associated virus 2 vector for gene therapy - Google Patents

Safe adeno-associated virus 2 vector for gene therapy Download PDF

Info

Publication number
CA2098483C
CA2098483C CA002098483A CA2098483A CA2098483C CA 2098483 C CA2098483 C CA 2098483C CA 002098483 A CA002098483 A CA 002098483A CA 2098483 A CA2098483 A CA 2098483A CA 2098483 C CA2098483 C CA 2098483C
Authority
CA
Canada
Prior art keywords
vector
aav
globin
gene
promoter
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.)
Expired - Fee Related
Application number
CA002098483A
Other languages
French (fr)
Other versions
CA2098483A1 (en
Inventor
Arun Srivastava
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.)
Research Corp Technologies Inc
Original Assignee
Research Corp Technologies Inc
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 Research Corp Technologies Inc filed Critical Research Corp Technologies Inc
Publication of CA2098483A1 publication Critical patent/CA2098483A1/en
Application granted granted Critical
Publication of CA2098483C publication Critical patent/CA2098483C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/795Porphyrin- or corrin-ring-containing peptides
    • C07K14/805Haemoglobins; Myoglobins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • 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
    • 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

Abstract

Gene therapy involves the transfer and stable insertion of new genetic information into cells. The present invention is directed to safe vectors for gene therapy and thus provides hybrid parvovirus vectors which are capable of site-specific integration into a mammalian chromosome without substantial cytotoxicity, and which direct erythroid cell-specific expression of heterologous genes. The hybrid vector is useful in gene therapy, particularly in the treatment of hemoglobinopathies and other hematopoietic diseases, and in conferring cell-specific multidrug resistance. A method of delivery of constitutive levels of a pharmaceutical product and a method of producing a recombinant protein are also provided.

Description

2 0 9 8 4 8 3 ~361Z.JMM
lm/spec/8361Z.FOR

Gene therapy involves the transfer and stable insertion of new genetic information into cells. The present invention is directed to a safe vector for gene therapy and provides hybrid parvovirus vectors which are capable of site-specific integration into a mammalian chromosome without substantial cytotoxicity, and which can direct cell-specific expression of a desired gene product. The hybrid vectors are useful in gene therapy, particularly in the treatment of hemoglobinopathies. A
method of delivery of a pharmaceutical product is also provided. The present invention also provides a method of conferring cell-specific multidrug resistance.
The therapeutic treatment of diseases and disorders by gene therapy involves the transfer and stable insertion of new genetic information into cells.
The correction of a genetic defect by re-introduction of the normal allele of a gene encoding the desired function has demonstrated that this concept is clinically feasible [Rosenberg _et _al. (1990) New Eng. J.
Med., 323, 570].
Hematopoietic stem cells or pluripotent progenitor cells are particularly useful for gene therapy studies since, although they are somatic cells, they differentiate to produce all the lineages of blood cells. Hence, the introduction of a foreign gene into a stem or progenitor cell results in the production of various lineages which can potentially express the foreign gene or alter control of native gene products.
The introduction of a foreign gene into a progenitor cell or any other appropriate cell requires a method of B

_2_ 2 0 9 8 4 8 3 1 gene transfer to integrate the foreign gene into the cellular genome. Although a variety of physical and chemical methods have been developed for introducing exogenous DNA into eukaryotic cells, viruses have generally been proven to be much more efficient for this purpose. Several DNA-containing viruses such as parvoviruses, adenoviruses, herpesviruses and poxviruses, and RNA-containing viruses, such as retroviruses, have been used to develop eukaryotic cloning and expression vectors. The fundamental problem with retroviruses is that they are either the etiologic agents of, or are intimately associated with, malignancy. Retroviruses integrate randomly into the cellular genome, and thus may activate cellular proto-oncogenes or may disrupt sequences critical to cell function. Accordingly, the use of retroviral vectors in gene transfer presents a problem in that there is a finite chance that such vectors may induce neoplasia.
Thus, a need exists for additional and improved vectors for gene transfer.
Whereas retroviruses are frequently the etiologic agents of malignant disorders, parvoviruses constitute the sole group of DNA-containing viruses that have not yet been associated with any malignant disease.
Although parvoviruses are frequently pathogenic in animals, a parvovirus of human origin, the adeno-associated virus 2 (AAV), has so far not been associated with any known human disease, even though up to 90~ of the human population has been exposed to AAV.
[Blacklow, N. R. (1988) in: Parvoviruses and Human Disease, CRC Press, Boca Raton]. In addition, most retroviruses used for gene transfer are of murine SUBSTITUTE SHEET

WO 93/09239 2 0 9 ~ ~. 8 3 P~T/US92/09769 1 origin, while AAV, a human virus, is physiologically more relevant for gene transfer in humans. Moreover, retroviruses are susceptible to inactivation by heat and organic solvents, whereas AAV is heat stable, extremely resistant to lipid solvents, and stable between pH 3.0 and 9Ø Thus as vehicles for gene transfer, parvoviruses provide many advantages over retroviruses.
Recombinant retroviruses have low viral titers (105-10~ virions/ml) (Rosenberg) in contrast to the high titers of recombinant AAV (lOB-109 virions/ml) [Srivastava et al. (1990) Blood _76, 1997].
Consequently, it is generally not possible to achieve an infection efficiency with recombinant retrovirus beyond 10-50°, of the target cell population, with successful infection requiring actively replicating cells. In contrast, a 70°s infection efficiency has been reported for a recombinant AAV [Samulski _et _al. (1989) J. Virol.
63, 3822], and it is possible to achieve a 100$
infectivity of target cells with wild-type AAV [Nahreini et al. (1989) Intervirol. 30, 74]. Furthermore, even though recombinant retroviral vectors have been rendered replication-incompetent, there remains a low probability of recombination between the vector and endogenous retroviral sequences. In contrast, 60-90~ of the population is sero-positive for human parvoviruses, and no endogenous viral sequences have yet to be detected in volunteer donors. In recombinant AAV vectors, all of the AAV coding sequences have, nonetheless, been deleted.
Perhaps the most significant advantages of AAV-based vectors are that they mediate integration into the host chromosomal DNA in a site-specific and stable SUBSTITUTE SHEET

20 9~ 48 3 manner. Retroviral genomes, following reverse transcription, undergo integration into the host chromosomal DNA with a totally random integration pattern. AAV establishes a latent infection which is site-specific. The integration site has been mapped to human chromosome 19. (Kotin et al. (1990) Proc. Natl.
Acad. Sci. USA 87, 2211). It has therefore become feasible to accomplish site-specific delivery of exogenous DNA into mammalian cells. While retroviral vectors mediate integration of non-viral sequences into the host chromosome, the integration pattern is not always stable. Frequently the integrated retroviral provirus is excised from the cell. AAV, on the other hand, establishes a stable integration.
Despite the potential advantages outlined above, the parvovirus-based vectors suffer from one limitation, and that is the size of a DNA sequence that can be packaged into the mature virions. For example, whereas up to 8.0 - 9.0 kilobase pair (kbp) DNA fragments can be packaged into retroviral vectors, a maximum of about 5.0 kbp DNA can be packaged into AAV. This size limitation, however, does not preclude the cloning and packaging of most cDNA molecules.
Thus parvovirus-based vectors offer a useful alternative to retroviral vectors for gene therapy in humans. While AAV-based vectors allow stable, site-specific integration of transferred genes, the indiscriminate expression of the transferred gene in all cell lineages presents significant problems. Thus, a need exists for AAV vectors which effect tissue-specific expression of the transferred gene. In accordance with the present invention, one method, for example, to solve SUBSTITUTE SHEET

_5_ 2098483 1 this problem is by a combination of the features of AAV
and another human parvovirus, B19.
While AAV causes no known disease, B19 is known to be the etiologic agent of a variety of clinical disorders in humans. B19 is the causative agent of transient aplastic crises associated with various hemolytic anemias, erythema infectiosum or the "fifth disease", post-infection polyarthralgia and thrombocytopenia in adults, and some cases of chronic bone marrow failure and hydrops fetalis.
AAV is dependent on a helper virus, such as adenovirus, herpesvirus, or vaccinia virus, for optimal replication. In the absence of a helper virus, AAV
establishes a latent infection in which the viral genome integrates into chromosomal DNA site-specifically. B19, on the other hand, is an autonomously replicating virus that is known to replicate only in human hematopoietic cells in the erythroid lineage. Both AAV and B19 contain linear, single-stranded DNA genomes, but their genomes show no homology at the nucleotide sequence level. The nucleotide sequences of both genomes are known. [Lusby et al. (1980) J. Virol. _34, 402, Srivastava et al. (1983) J. Virol. _45, 555; Shade _et _al.
(1986) J. Virol. 58, 921]. The AAV genome contains inverted terminal repeats (ITRs) of 145 nucleotides, 125 nucleotides of which form a palindromic hairpin that plays a critical role during AAV DNA replication. The sequences of the ITRs are shown in Fig. 1 and as SEQ ID
No:l. In latently infected cells, the termini of AAV
are at the junction of the cellular sequences and thus the termini also facilitate integration and rescue.
SUBSTITUTE SHEET

2o~s~-s3 The remarkable features of the two human parvoviruses can be combined, for example, in an AAV-B19 hybrid vector, to provide vectors in accordance with the present invention. The vectors of this invention are particularly useful for gene transfer in bone marrow cells and other hematopoietic cells. These hybrid viral vectors mediate site-specific integration as well as tissue-specific expression of heterologous genes in hematopoietic cells.
The present invention is directed to hybrid parvovirus vectors capable of site-specific integration into a mammalian chromosome without substantial cytotoxicity, and which can direct tissue-specific expression of a heterologous gene, i.e. a non-parvovirus gene. More particularly, the present invention provides vectors comprising two inverted terminal repeats of adeno-associated virus 2 and at least one genetic cassette comprising a promoter capable of effecting cell-specific expression operably linked to a heterologous gene wherein the cassette resides between the two inverted terminal repeats. In a preferred embodiment, the promoter is the p6 promoter of B19 parvovirus and directs erythroid cell-specific expression of~the heterologous gene.
In another aspect of this invention, host cells transduced by the hybrid vectors of the present invention are provided.
Another aspect of the present invention provides a method of treatment for hematopoietic diseases, in particular hemoglobinopathies, by transducing hematopoietic stem or progenitor cells with a vector of a present invention and introducing the transduced cells SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ ~ g 3 PCT/US92/09769 1 into a patient, where the heterologous gene is expressed.
A further aspect of this invention provides a method for delivery of a pharmaceutical product in a mammal by transducing hematopoietic stem or progenitor cells with a Hybrid vector of the present invention and introducing the transduced cells into the mammal. The heterologous gene is expressed, and the mature red blood cell provides a vehicle for delivery of the heterologous gene product throughout the bloodstream or to the liver or spleen.
Yet another aspect of the present invention provides a method of conferring cell-specific multidrug resistance by transducing cells with a hybrid vector of the present invention in which the heterologous gene is a multidrug resistance gene, and introducing the transduced cells into a mammal. In a preferred embodiment, the hybrid vector contains the B19p6 promoter, and thus the multidrug resistance phenotype is conferred to erythroid cells.
As used herein, transduction refers to a process by which cells take up foreign DNA and integrate that foreign DNA into their chromosomes. Transduction can be accomplished, for example, by transfection, which refers to various techniques described hereinbelow by which cells take up DNA, or infection, by which viruses are used to transfer DNA into cells.
Fig. 1 depicts the nucleotide sequence of an ITR
of the AAV 2 genome.
Fig. 2 depicts the nucleotide sequence of B19 from nucleotide number 200 to nucleotide number 424 as numbered by Shade et al. (1986).
SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ ~ PCT/US92/09769 _g_ 1 Fig. 3 diagrams the construction of a hybrid vector of the present invention.
Fig. 4 demonstrates the site-specific integration of the wild-type AAV genome into normal human diploid chromosomal DNA by Southern blot analysis.
Fig. 5 demonstrates the site-specific integration of the recombinant AAV genome into normal human bone marrow cells by Southern blot analysis.
Fig. 6 is a diagram of the construction of recombinant plasmids pWP-7A (Panel A) and pWP-19 (Panel B).
Fig. 7 is a diagram of the construction of recombinant plasmid pWN-1.
Fig. 8 is a diagram of the construction of recombinant plasmids pWP-21 and pWP-22 (Panel A) and pWP-16 and pWP-17 (Panel B).
Fig. 9 demonstrates Southern blot analysis of rescue and replication of the recombinant neon gene in human cells. Panel A: Rescue from plasmid pWP-8A;
Panel B: Rescue from plasmid pWP-21; Panel C: Rescue from plasmid pWP-22; Panel D: Rescue from plasmid pWP-16; Panel E: Rescue from plasmid pWP-17. Recombinant plasmids were transfected separately in adenovirus-infected Human KB cells (Lanes 1, 3, 5, 7, 9), or co-transfected with pAAV/Ad helper plasmid (Lanes 2, 4, 6, 8, 10). m and d denote the monomeric and dimeric forms, respectively, of the recombinant AAV DNA replicative intermediates.
Fig. 10 is a diagram of the construction of a hybrid vector in which the neomycin resistance (Neon) gene is under the control of the B19p6 promoter.
SUBSTITUTE SHEET

_g_ 1 Fig. 11 is a graph depicting cell viability after AAV-mediated transfer of Neor to human hematopoietic stem cells. Panel A illustrates Neor gene expression under the control of the TK promoter. Panel B
illustrates Neor gene expression under the control of the B19p6 promoter.
Fig. 12 depicts recombinant AAV vectors of the present invention containing selectable genes under the control of B19p6 promoter and a human erythroid-specific enhancer (HS-2).
Fig. 13 depicts the recombinant AAV vectors of the present invention which express the human f3-globin gene and the Neor gene.
Fig. 14 provides a Souther blot of DNA isolated from vHS2/b-globin-Neo infected K562 cells.
Fig. 15 provides a Northern blot of RNA from K562 cells infected with vHS2/b-globin-Neo and vHS2/B19-globin-Neo.
The present invention relates to hybrid parvovirus vectors which comprise a pair of AAV inverted terminal repeats (ITRs) which flank at least one cassette containing a promoter which directs cell-specific expression operably linked to a heterologous gene. Heterologous in this context refers to any nucleotide sequence or gene which is not native to the AAV or B19 parvovirus. In accordance with the present invention, AAV and B19 coding regions have been deleted, resulting in a safe, nvncytotoxic vector.
Representative heterologous genes are described hereinbelow. The AAV ITRs, or modifications thereof, confer infectivity and site-specific integration, but not cytotoxicity, and the promoter directs cell-specific SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ ~ ~ PCT/US92/09769 1 expression and preferably erythroid cell expression using the p6 promoter of B19 parvovirus. The hybrid vectors of the present invention thus provide DNA
molecules which are capable of integration into a mammalian chromosome without substantial toxicity.
These hybrid vectors allow safe integration of the DNA
into the cellular genome, since the portions of the DNA
responsible for replication of the parvovirus have been deleted, and therefore these vectors cannot self replicate.
In accordance with the present invention, the hybrid vector comprises a first and second terminal repeat which flank a promoter linked to a heterologous gene. The terminal repeats can comprise all or part of the ITRs of AAV. The terminal repeats mediate stable integration of the DNA sequence into a specific site in a particular chromosome, e.g. human chromosome 19. The entire DNA sequence, including the ITRs, the promoter, and the heterologous gene can be integrated into the cellular genome.
The terminal repeats of the hybrid vector of the present invention can be obtained by restriction endonuclease digestion of AAV or a plasmid such as psub201, which contains a modified AAV genome [Samulski et al. (1987) J. Virol. 61, 3096], or by other methods known to the skilled artisan, including but not limited to chemical or enzymatic synthesis of the terminal repeats based upon the published sequence of AAV. The ordinarily skilled artisan can determine, by well-known methods such as deletion analysis, the minimum sequence or part of the AAV ITRs which is required to allow function, i.e. stable and site-specific integration.
SUBSTITUTE SHEET

1 The ordinarily skilled artisan can also determine which minor modifications of the sequence can be tolerated while maintaining the ability of the terminal repeats to direct stable, site-specific integration. Site-specific integration can be assessed, for example, by Southern blot analysis. DNA is isolated from cells transduced by the vectors of the present invention, digested with a variety of restriction enzymes, and analyzed on Southern blots with an AAV-specific probe. A single band of hybridization evidences site-specific integration.
Other methods known to the skilled artisan, such as polymerase chain reaction (PCR) analysis of chromosomal DNA can be used to assess stable integration.
The vectors of the present invention contain a promoter which directs tissue-specific expression. For example, the wild-type parvovirus B19 has a limited host range and exhibits a remarkable tissue tropism for the erythroid elements of bone marrow. In a preferred embodiment, the hybrid vectors of the present invention utilize a transcriptional promoter of B19 to effect tissue-specific expression of heterologous sequences.
In a more preferred embodiment the promoter is the p6 promoter of B19, which is active in erythroid progenitor cells. The nucleotide sequence of B19 from nucleotide number 200 to nucleotide number 424 as numbered by Shade et al. (1986) contains the p6 promoter and is depicted in Fig. 2 and as SEQ ID N0:2.
The consensus promoter-like sequence TATATATA is present at nucleotide 320 in B19 (as numbered by Shade et al.) and thus transcription is likely to originate about 30 nucleotides downstream. It has been discovered in accordance with the present invention that B19 SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ 4 ~ ~ PCT/US92/09769 fragments containing these sequences direct expression that is specific for erythroid progenitor cells, and that deletion of B19 coding sequences downstream from the promoter prevents replication of B19. As explained above, one of ordinary skill in the art can determine the minimum sequence and modifications of the p6 promoter which provide cell-specific, non-cytotoxic expression. This can be determined by infecting erythroid and non-erythroid cells with vectors containing the B19p6 promoter and assessing expression of the heterologous gene. The promoter sequence can be derived by restriction endonuclease digestion of B19 or a cloned B19 plasmid such as pYT103 and pYT107 [Cotmore et al. (1984) Science 226, 1161] or by any other methods known to the skilled artisan, including but not limited to chemical or enzymatic synthesis based upon the published sequence of B19. Other cell-specific promoters can be obtained by analogous methods, and the specificity of these promoters is determined by assessing expression in the appropriate cell type.
The promoter of the hybrid vector is operably linked to the heterologous gene. Any gene that can be transcribed in such a construction is contemplated by the present invention. In a preferred embodiment, the heterologous gene encodes a biologically functional protein, i.e. a polypeptide or protein which affects the cellular mechanism of a cell in which the biologically functional protein is-expressed. For example, the biologically functional protein can be a protein which is essential for normal growth of the cell or for maintaining the health of a mammal. The biologically functional protein can also be a protein which improves SUBSTtTU i E SHEET

2_Q~9~483 1 the health of a mammal by either supplying a missing protein, by providing increased quantities of a protein which is underproduced in the mammal or by providing a protein which inhibits or counteracts an undesired molecule which may be present in the mammal. The biologically functional protein can also be a protein which is a useful protein for investigative studies for developing new gene therapies or for studying cellular mechanisms.
The biologically functional protein can be a protein which is essential for normal growth or repair of the human body. The biologically functional protein may also be one which is useful in fighting diseases such as cancer, atherosclerosis, sickle-cell anemia and the thalassemias. Examples of such biologically functional proteins are hemoglobin (a, a or ~y-globin), hematopoietic growth factors such as granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), granulocyte colony stimulating factor (G-CSF) and erythropoietin (EPO). Another example is tumor necrosis factor (TNF), which is a molecule that can be used to treat cancer, and in particular, tumors. The tumor suppressors p53 and retinoblastoma (RH) are also contemplated. Various cytokines such as mast cell growth factor (MGS) and interleukins 1-11 are also proteins which are contemplated by the present invention. The biologically functional protein may also be a selectable marker for antibiotic resistance such as a selectable marker for neomycin resistance in eukaryotes. Other types of selectable markers such as adenine phosphoribosyl transferase (APRT) in APRT-SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ PCT/US92/09769 1 deficient cells, or the firefly luciferase gene are also included. The heterologous genes encoding these proteins can be provided by any of a variety of methods, such as routine cloning procedures (Sambrook et al.), excision from a vector containing the gene of interest, or chemical or enzymatic synthesis based on published sequence information. In many instances the DNA
encoding the protein of interest is commercially available.
The biologically functional protein can affect cellular mechanism by providing a new or altered function to a cell. For example, the heterologous gene can be a multidrug resistance gene (mdr) which encodes P-glycoprotein. P-glycoprotein is a cell membrane glycoprotein which affects intracellular drug accumulation and is responsible for the phenomenon of mutidrug resistance.(for review, see Biedler [1992]
Cancer 70 1799) In another embodiment the heterologous gene can encode a non-biologically functional protein. For example, a hybrid gene comprising various domains and functions from a variety of sources can be designed and produced by recombinant technology or enzymatic or chemical synthesis.
In another preferred embodiment the heterologous gene is capable of being transcribed into an RNA
molecule which is sufficiently complementary to hybridize to an mRNA or DNA of interest. Such an RNA
molecule is hereinafter referred to as antisense RNA, 3p and has utility in preventing or limiting the expression of overproduced, defective, or otherwise undesirable molecules. The vector of the present invention can SlIBSTiTIlTE SHEET

WO 93/09239 ~ ~ ~ ~ '~ ~ 3 PCT/US92/09769 1 comprise, as the heterologous gene, a sequence encoding an antisense RNA which is sufficiently complementary to a target sequence such that it binds to the target sequence. For example, the target sequence can be part of the mRNA encoding a polypeptide such that it binds to and prevents translation of mRNA encoding the polypeptide. In another embodiment, the target sequence is a segment of a gene that is essential for transcription such that the antisense RNA binds the segment (e. g. a promoter or coding region) and prevents or limits transcription. Hence, the antisense RNA must be of sufficient length and complementarity to prevent translation of its target mRNA or transcription of its target DNA.
In a preferred embodiment the antisense RNA is a l5mer and exhibits 100°s complementarity to the target sequence. One of ordinary skill in the art can determine longer or shorter antisense molecules having sufficient complementarity to a target sequence such that the antisense molecule is capable of binding to the target and thereby inhibiting translation or transcription. The heterologous gene can be provided, for example, by chemical or enzymatic synthesis, or from commercial sources.
It is preferable that the length of the heterologous gene is such that the overall size of the hybrid vector is about 5 kilobases (kb), since the packaging limit of AAV virions is about 5 kb (Hermonat et al. (1984) Proc. Natl. Acad. Sci. USA 81, 6466).
The hybrid vectors of the present invention can be provided by inserting the heterologous gene and the cell-specific promoter between a pair of AAV-derived SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ I~g ~ PCT/US92/09769 1 terminal repeats. The combination of a promoter and heterologous gene is also referred to herein as a cassette. Thus, the invention provides a vector in which: 1) the terminal repeats mediate stable, site-s specific integration into the cellular genome; and 2) the promoter mediates cell-specific expression of a heterologous gene, e.g. in erythroid cells, or the promoter mediates transcription of an antisense RNA or a sense RNA encoding a polypeptide of interest. The promoter sequence is operably linked to the heterologous gene in a manner to effect expression of the gene.
Hence, the promoter sequence can be at either or both ends of the heterologous sequence or coding region.
Furthermore, more than one promoter and heterologous gene can be present in one vector, i.e. there can be two or more cassettes between the ITRs. Accordingly, more than one heterologous gene can be expressed by one vector.
Standard techniques for the construction of such hybrid vectors are well-known to those of ordinary skill in the art and can be found in references such as Sambrook et al. (1989) in Molecular Cloning: A
Laboratory Manual, Cold Spring Harbor, New York, or any of the myriad of laboratory manuals on recombinant DNA
technology that are widely available. A variety of strategies are available for ligating fragments of DNA, the choice of which depends on the nature of the termini of the DNA fragments and can be readily determined by the skilled artisan.
It is further contemplated in accordance with the present invention to include in the hybrid vectors other nucleotide sequence elements which facilitate suas~rrruTt s~EEr WO 93/09239 2 p g g 4 g 3 PCT/LJS92/09769 1 integration of DNA into chromosomes, expression of the DNA, and cloning of the vector. For example, the presence of enhancers upstream of the promoter or terminators downstream of the coding region can facilitate expression. In another example, recent studies have identified a DNasel-hypersensitive site (HS-2) upstream of the human globin gene cluster that significantly enhances the erythroid-specific expression of the globin genes. [Tuan et al. (1985) Proc. Natl.
Acad. Sci. USA 82, 6384]. In the hybrid vectors of the present invention, the presence of HS-2 upstream of the B19p6 promoter enhances tissue specific expression.
As described hereinabove, the vectors of the present invention can be constructed by a variety of well-known methods, and the order of the ligation of the elements can be varied. In a preferred embodiment the cell-specific promoter and heterologous gene are ligated together to provide a cassette which can be inserted between two AAV-ITRs. For example, to provide a cassette containing the B19p6 promoter and a heterologous gene, a fragment containing the p6-promoter is inserted into a pUCl9 plasmid, after which the p6 containing plasmid is linearized by restriction enzyme cleavage downstream of the p6 promoter. The heterologous gene is then inserted immediately downstream of the p6 promoter. A fragment containing both the p6 promoter and the heterologous gene is excised from the plasmid and inserted between the AAV-ITRs in an AAV plasmid from which the AAV coding regions have been deleted. The resulting plasmid comprises the p6 promoter and a heterologous gene flanked by a pair of AAV-ITRs. This construction is described more SUBSTITUTE SHEET

WO 93/09239 2 0 9 ~ 4 ~ 3 PCT/US92/09769 1 specifically as follows and is diagrammed in Fig. 3. To generate a plasmid containing p6, a fragment containing the p6 promoter of B19 is isolated from B19 DNA or cloned B19 DNA [see, for example, Cotmore et al. (1984);
Shade et al. (1986)]. In a preferred embodiment this B19 fragment corresponds to nucleotides 200 to 480 as numbered by Shade et al. (1986) and contains the entire 5' non-coding region and p6 promoter of B19. This 280 by fragment is flanked by EcoRI and XbaI restriction sites and can be generated by cleavage with these restriction enzymes. This fragment is cloned into the EcoRI-XbaI sites of pUCl9 to generate plasmid pB19p6.
The skilled artisan will recognize that other plasmids and restriction sites can be utilized to generate a vector comprising a B19p6 promoter. Alternatively, the B19p6 promoter can be synthesized chemically or enzymatically based upon the published sequence and ligated to the heterologous gene.
A heterologous gene can be operably linked downstream of the B19p6 promoter fragment as follows.
The plasmid pB19p6 is cleaved with HincII, which cleaves B19 DNA downstream of the p6 promoter (i.e. at nucleotide 424) and also in the multiple cloning site of pUCl9. The desired heterologous gene is blunt-end ligated downstream from the B19p6 promoter between the two HincII sites to generate a plasmid pB19p6-insert.
The ordinarily skilled artisan will recognize a variety of methods, as exemplified, e.g. in Sambrook et al.
(1989), to ligate a fragment containing a cell-specific promoter with a fragment containing the heterologous gene. In accordance with the present invention, the coding sequence of GM-CSF, APRT, neon, the SUBSTITUTE SHEET

1 retinoblastoma gene, a-globin, Li-globin and y-globin have been employed as the heterologous gene, resulting in the construction of hybrid vectors, designated AAV-B19-GM-CSF, AAV-B19-APRT, AAV-B19-neo=, AAV-B19-RH, AAV-H19-a-globin, AAV-B19-f3-globin and AAV-H-19-globin, respectively. A multidrug resistance gene encoding P-glycoprotein is also specifically contemplated as the heterologous gene. The coding sequences of the respective genes are known [Lee _et _al. (1985) Proc.
Natl. Acad. Sci. USA 82, 4360 (GM-CSF); Broderick _et _al.
(1987) Proc. Natl. Acad. Sci. USA 84, 3349 (APRT);
Tratschin et al. (1985) Mol. Cell. Biol. 5, 3251 (Neon);
Huang et al. (1988) Science 242, 1563 (RH-1); Liebhaber et al. (1980) Proc. Natl. Acad. Sci. USA _77, 7054 (a-globin); Lawn et al. (1980) Cell 21, 647 (~i-globin);
Enver et al. (1989) Proc. Natl. Acad. Sci. USA _86, 7033 (y-globin) Roninson et al. (1986) Proc. Nat'1 Acad. Sci.
USA 83, 4538; Roninson et al. (1991) in Molecular and Cellular Biology of Multidrua Resistance in Tumor Cells (ed. Roninson, Plenum Press, NY) 91-106; Schinkel _et _al.
(1991) Cancer Res. 51, 2628; Chen _et _al. (1990) J. Biol.
Chem. 265, 506; (mdr)] and thus can be easily provided as described hereinabove.
The pB19p6-insert plasmid exemplifies a promoter-heterologous gene cassette which can be isolated by digesting the plasmid, for example, with EcoRI and HindIII or other appropriate restriction enzymes, and then ligated between two AAV-ITRs. The AAV-ITRs are provided by, for example, restriction digestion of AAV
DNA or AAV cloned DNA, or chemical or enzymatic synthesis based upon the published sequence of AAV ITRs [Lusby et al. (1980)]. In a preferred embodiment, the SUBSTITUTE SHEET

~i~J~483 1 AAV ITRs comprise the 145 nucleotides shown in Fig. 1.
Fragments which contain the 125 nucleotides which form the palindromic hairpin (nucleotide 1-125 of Fig. 1) or longer fragments which contain the terminal 191 nucleotides of the viral chromosome are also useful.
Additional endogenous sequences, for example linkers to facilitate cloning and ligation, can also be used in the constructs. In a preferred embodiment, the AAV ITRs are provided by a plasmid, e.g. psub201 [Samulski et al.
(1987)] which is an AAV derivative into which XbaI
cleavage sites have been introduced at sequence positions 190 and 4484, and the right-terminal 191 base pairs of the viral genome have been substituted for the normal left-terminal 190 base pair domain. This modification results in the extension of the psub201 terminal repeats to 191 base pairs. The XbaI cleavage sites allow substitution of the AAV coding region with exogenous sequences, i.e. the B19 promoter and heterologous gene, such that the exogenous sequences are flanked by the AAV-ITRs. Derivatives of psub201 engineered to contain other restriction sites, as demonstrated in Examples 2 and 3, are also useful for providing the AAV-ITRs.
To substitute the B19p6-insert, i.e. the cassette, for the AAV coding region, psub201 is digested with Xbal to delete the AAV coding regions. Plasmid vector DNA containing the AAV-ITRs is isolated and ligated to the B19p6-insert construction. Ligation may be facilitated by the addition of adapters to the AAV-ITRs and linkers to the B19-p6-insert.
For example, in another preferred embodiment the vectors of the present invention are constructed by SUBST~tUTE SHEET

WO 93/09239 ~ ~ ~ ~ ~~ ~ PCT/US92/09769 1 modifying the engineered XbaI sites of psub201 to provide additional restriction sites, deleting the AAV
coding regions by digestion with the appropriate restriction enzyme and ligating the B19p6-insert cassette to the plasmid DNA containing the AAV-ITRs.
The construction of prototype plasmid vectors containing the AAV-ITRs but not the AAV coding regions, and which further contain cloning sites to facilitate the insertion of promoter-heterologous gene cassettes, are exemplified in Examples 2 and 3.
The resulting plasmid comprises a cell-specific promoter upstream of a heterologous sequence, both of which are flanked by AAV-ITRs. The order of the ligations, the nature of the complementary ends, the use of linkers and adapters, and other details can be varied as necessary by one of ordinary skill in the art to provide the AAV-B19 hybrid vector of the present invention.
To establish integration of the vector DNA into the chromosome of a host cell, host cells are transfected with the vector or infected with mature virions containing the hybrid vectors. Methods of DNA
transfection are well-known to one of ordinary skill in the art and include, for example, naked DNA
transfection, microinjection and celY fusion. More efficient integration is accomplished by infection with virions containing the hybrid vectors.
Virions can be produced by coinfection with a helper virus such as adenovirus, herpes virus or vaccinia virus. Following coinfection of host cells with the subject vector and a helper virus, virions are isolated and the helper virus is inactivated. The SUBSTITUTE SHEET

~U98483 1 resulting helper free stocks of virions are used to infect host cells. In another embodiment, virions are produced by cotransfecting helper virus-infected cells with the vector of the present invention and a helper plasmid. For example, the hybrid construct of the present invention can be packaged into mature AAV
virions by cotransfection of adenovirus-infected cells with the vector of the present invention and a plasmid which provides the parvovirus red gene and adenovirus termini. An example of such a plasmid is pAAV/Ad, which contains the entire coding sequence of AAV and the adenovirus type 5 terminal sequences in place of the normal AAV termini. [Samulski et al. (1989)].
Following cotransfection, mature virions are isolated by standard methods, e.g. cesium chloride centrifugation and heated at 56°C for our hour to inactivate any contaminating adenovirus. The resulting mature virions contain the vector of the present invention and are used to infect host cells in the absence of helper virus.
Function of the hybrid vectors of the present invention, i.e. the ability to mediate transfer and expression of the heterologous gene in a specific cell type, can be evaluated by monitoring the expression of the heterologous gene in transduced cells. For example, bone marrow cells are isolated and enriched for hematopoietic stem cells (HSC), e.g. by fluorescence activated cell sorting as described in Srivastava et al.
(1988) J. Virol. 62, 3059. HSC are capable of self-renewal as well as initiating long-term hematopoiesis and differentiation into multiple hematopoietic lineages in vitro. HSC are transfected with the vector of the present invention or infected with varying SUBSTITUTE SHEET

WO 93/09239 z Q ~ ~ 4 g ~ PCT/US92/09769 1 concentrations of virions containing a subject hybrid vector and then assessed for the expression of the heterologous gene.
The assay for expression depends upon the nature of the heterologous gene. Expression can be monitored by a variety of methods including immunological, histo-chemical or activity assays. For example, Northern analysis can be used to assess transcription using appropriate DNA or RNA probes. If antibodies to the polypeptide encoded by the heterologous gene are available, Western blot analysis, immunohistochemistry or other immunological techniques can be used to assess the production of the polypeptide. Appropriate biochemical assays can also be used if the heterologous gene is an enzyme. For example, if the heterologous gene encodes antibiotic resistance, a determination of the resistance of infected cells to the antibiotic can be used to evaluate expression of the antibiotic resistance gene.
In addition to assessing that the heterologous gene is expressed in the appropriate cells, the correct promoter specificity of the hybrid vectors can be evaluated by monitoring the expression of the heterologous gene, or lack of expression, in cells in which the promoter is not expected to be active. For example, when cells from a naso-pharyngeal cell line, KB, are transduced with a hybrid vector containing the B19p6 promoter, the heterologous gene is not expressed, since the B19p6 promoter is erythroid cell-specific.
Detection of the heterologous gene product at levels at or below the level of untransduced cells confirms that the B19p6 promoter of the hybrid vector does not direct SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ c~ ~ ~ ~ PCT/US92/09769 1 expression of the heterologous gene in non-hematopoietic cells.
The hybrid vectors of the present invention are useful for gene therapy. In particular, the vectors of the present invention can direct erythroid cell-specific expression of a desired gene, and thus are useful in the treatment of hemoglobinopathies.
It is contemplated in accordance with the present invention to use the hybrid vector in the treatment of a variety of diseases, including thalassemia, sickle-cell anemia, diabetes and cancer. The heterologous gene can be the normal counterpart of one that is abnormally produced or underproduced in the disease state, for example a-globin for the treatment of sickle-cell anemia, and a-globin, !3-globin or Y-globin in the treatment of thalassemia. The heterologous gene can encode antisense RNA as described hereinabove. For ' example, a-globin is produced in excess over B-globin in 13-thalassemia. Accordingly, B-thalassemia can be treated in accordance with the present invention by gene therapy with a vector in which the heterologous gene encodes an antisense RNA. The antisense RNA is selected such that it binds to a target sequence of the a-globin mRNA to prevent translation of a-globin, or to a target sequence of the a-globin DNA such that binding prevents transcription of a-globin DNA. In the treatment of cancer the heterologous gene can be a gene associated with tumor suppression, such as retinoblastoma gene, the anti-oncogene p53, or the gene encoding tumor necrosis factor.
The use of the hybrid vectors of the present invention for the treatment of disease involves SUBSTITUTE SHEET

-25- 2 0 9 8~4 8 3 1 transduction of HSC or progenitor cells with the hybrid vector. Transduction is accomplished by transfection with the vector or preparation of mature virions containing the hybrid vectors and infection of HSC or progenitor cells with the mature virions. Transduced cells are introduced into patients, e.g. by intravenous transfusion (see, for example, Rosenberg, 1990). HSC or progenitor cells are provided by obtaining bone marrow cells from patients and optionally enriching the bone marrow cell population for HSC. HSC can be transduced by standard methods of transfection or infected with mature virions for about one to two hours at about 37°C.
Stable integration of the viral genome is accomplished by incubation of HSC at about 37°C for about one week to about one month. The stable, site-specific integration and erythroid cell-specific expression is assessed as described above. After the transduced cells have been introduced into a patient, the presence of the heterologous gene product can be monitored or assessed by an appropriate assay for the gene product in the patient, for example in peripheral red blood cells or bone marrow of the patient when expression is erythroid cell-specific. As described hereinabove, the specific assay is dependent upon the nature of the heterologous gene product and can readily be determined by one skilled in the art.
For example, 13-thalassemia represents a heterologous group of clinical syndromes that are inherited as mutated alleles of genes that encode the human ti-globin chain. These mutations affect all aspects of f3-globin gene expression including transcription, splicing, polyadenylation, translation SUBSTITUTE SHEET

2 0 9 8 ~ 8 3 -26-1 and protein stability. The hallmark of 13-thalassemia is the marked reduction or total absence of synthesis of normal adult hemoglobin (HbA; aztiz). Despite significant advances in the understanding of basic underlying molecular mechanisms of f3-thalassemia, treatment is limited to regular red blood cell transfusions and iron-chelation therapy. Treatment by bone marrow transplantation has also been attempted [Thomas et al. (1982) Lancet, ii, 227], but an effective cure has not been found.
Accordingly,. the vectors of the present invention are useful in the treatment of 13-thalassemia. An AAV-B19 vector is constructed in which the heterologous gene is the normal human b-globin gene, with the resulting AAV-B19-Q-globin vector allowing parvovirus-mediated transfer, site-specific integration and erythroid cell-specific expression of the normal human Ci-globin gene in human hematopoietic cells.
Abnormal 13-globin expression Li-thalassemia may result in the overabundance of a-globin mRNA relative to f3-globin mRNA. The present invention cannot only provide a normal b-globin gene, as described hereinabove, but can further be utilized to down-regulate the production of excess a-globin by providing a vector with an antisense RNA as the heterologous gene.
An AAV-B19 hybrid vector is constructed in which the heterologous sequence encodes an antisense RNA which is sufficiently complementary to a region of the mRNA
encoding the a-chain, such that it binds to and prevents translation of the a-globin mRNA, or to a region of the DNA encoding a-globin such that it binds to and prevents transcription of the a-globin gene. Hence, the present SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ ~ ~ ~ PCT/US92/09769 1 invention contemplates gene therapy for B-thalassemia comprising transduction of hematopoietic stem or progenitor cells with a hybrid vector encoding normal B-globin chains, or simultaneous transduction with a vector encoding a normal B-globin chain and a vector encoding an RNA antisense to a-globin mRNA or DNA.
Alternately, a construction with more than one B19p6 promoter, as described hereinabove, permits coincident expression of B-globin and antisense a-globin.
Accordingly, transduction with a single vector effects both the provision of a normal B-globin gene and the down-regulation of excess a-chains. More specifically, bone marrow cells are transfected with the subject vectors, and transduced cells are introduced, e.g. by intravenous transfusion, into a patient. The stable integration of the vector can be assessed by PCR or Southern blot analysis and the expression of the heterologous gene can be evaluated by assaying for the heterologous gene product in the patient's peripheral blood cells or bone marrow cells. As described previously, the particular assay depends upon the nature of the heterologous gene product.
The vectors of the present invention are also useful in conferring cell specific multidrug resistance.
An AAV vector is contructed to contain a cell-specific promoter and a multidrug resistance gene, with the resulting vector allowing parvovirus-mediated transfer, site-specific integration and cell-specific expression of the mdr gene in a selected cell type. In a preferred embodiment, the vector is AAV-B19-mdr, and confers the multidrug resistance phenotype to erythroid cells.
SUE~STtTi~TE SHEET

~~ ~g~ _2g_ 1 Any of the numerous mdr genes known to confer the mdr phenotype is useful as the heterologous gene. The mdr genes are known to the ordinarily skilled artisan and are described, for example, by Roninson et al.
(1991) in Molecular and Cellular Biology of Multidrug Resistance in Tumor Cells (ed. Roninson, Plenum Press, NY) 91-106. Accordingly, the present invention provides a method of conferring cell-specific multidrug resistance which comprises transducing cells with the hybrid vector of the present invention which contains a cell-specific promoter and an mdr gene. In a preferred embodiment, the present invention provides a method of conferring multidrug resistance to erythroid cells of a patient which comprises obtaining bone marrow or stem cells from a patient, transducing the bone marrow or stem cells with the AAV-B19 hybrid vector of the present invention which contains an mdr gene as the heterologous gene, and reintroducing the transduced cells into a patient. The expression of the mdr gene can be evaluated by assaying for the mdr gene product, i.e., P-glycoprotein, in the patient's peripheral blood cells or bone marrow cells. For example, P-glycoprotein can be detected by known immunologic assays with an antibody against P-glycoprotein. Such antibodies are known and available to the ordinarily skilled artisan, and are described, for example, by Meyers et al. (1989) Cancer Res. 49, 3209 and Georges et al. (1990) Proc. Nat'1 Acad. Sci. USA 87, 152. The present method is particularly useful as an adjunct to cancer chemotherapy in that it effectively results in the protection of erythroid cells from the effects of chemotherapeutic agents targeted to other tissues.
SUBSTITUTE SHEET

1 Yet another aspect of the present invention provides a method for delivery of a pharmaceutical product, a protein or an antisense RNA in a mammal.
Since the normal differentiation of these stem cells results in production of mature erythrocytes, the transduction of stem cells with the subject vector ultimately yields a population of circulating, enucleate vesicles containing the gene product. This method comprises transducing hematopoietic stem or progenitor cells with the hybrid vector of the present invention and introducing, e.g. by intravenous transfusion or injection, the transduced cells into a mammal.
Transduction can be accomplished by transfecting cells with the hybrid vector by standard methods of infecting cells with mature AAV virions containing the hybrid vector at about 37°C for about one to two hours. Stable integration of the recombinant viral genome is accomplished by incubating cells at about 37°C for about one week to about one month. Transduced cells are recognized by assaying for expression of the heterologous gene, as described hereinabove. In this embodiment, the pharmaceutical product is encoded by the heterologous gene of the hybrid vector, and can be any pharmaceutical product capable of being expressed by the hybrid vector. Such products include a, b and y-globin, insulin, GM-CSF, M-CSF, G-CSF, EPO, TNF, MGF, interleukins, the gene product of the retinoblastoma gene, p53 or adenosine deaminase. Therefore, the present invention can provide production of constitutive levels of heterologous gene products inside membrane vesicles, specifically red blood cells, for in situ treatment of disease. Optionally, the hybrid vector can SiJE3ST~TUT~ St~EET

20 9848 3 ~g~-1 further comprise a sequence which encodes a signal peptide or other moiety which facilitates the secretion of the gene product from the erythroid cell. Such sequences are well-known to one of ordinary skill in the art [see, for example, Michaelis et al. (1982) Ann. Rev.
Microbiol. 36, 435] and can be inserted into the subject vectors between the promoter and coding region by methods described herein. This method can be used to treat a variety of diseases and disorders and is not limited to the treatment of hemoglobinopathies, since the heterologous gene is constitutively expressed and can be released from the red blood cell by virtue of a secretory sequence, or released when red blood cells are lysed in the liver and spleen.
The following examples further illustrate the present invention.

SUBSTITUTE SHEET

Site-Specific Inte ration of the Recombinant AAV Genome Site-specific integration of the AAV genome was confirmed by an approach in which normal human diploid fibroblasts (HDF) were either mock-infected, or infected with an increasing multiplicity-of-infection (moi) of wild-type AAV. Following multiple serial passage of these cells in culture, their total genomic DNA was isolated, digested with a variety of restriction endonucleases, and analyzed on Southern blots using an AAV-specific DNA probe. A representative Southern blot is presented in Fig. 4. Restriction enzymes are indicated at the top of the figure. The moi is indicated at the top of each lane, with 0.0 indicating mock-infection. The predominant single band of hybridization is evidence that the wild-type AAV genome integrates into normal human diploid cell chromosomal DNA in a site-specific manner. The target site was saturated only at very high moi of AAV, and no selection procedure was employed to select for cell populations that have the integrated provirus.
The site-specific integration of the recombinant AAV genome is demonstrated utilizing human bone marrow cells, which are the target cells for therapy of hemoglobinopathies.
Bone marrow cells were obtained from hematologically normal volunteer donors, and low-density, mononuclear bone marrow (LDBM) cells were isolated by Ficoll-Hypaque density centrifugation. LDBM
cells were infected with the recombinant AAV-Neo virions SUBSTITUTE SHEET

2Q 98 ~8 3 1 (vSV40-Neo), in which the Neo gene, under the control of the SV40 early promoter, is encapsidated into AAV
particles, and incubated in the presence of various cytokines such as GM-CSF (1 ng/ml) and IL-3 (1 ng/ml) for 48 hours. The cells were incubated in liquid cultures in the presence of 6418 at 37°C for 10 days, their total genomic DNA was isolated, cleaved with BamHI, and analyzed on a Southern blot using a Neo-specific DNA probe as shown in Fig. 5. Concentration of 6418 is indicated at the top of each lane. The single band of hybridization indicated by the arrow demonstrates that the recombinant AAV viral genome undergoes site-specific integration into human bone marrow cell chromosomal DNA.

SUBSTITUTE SHEET

WO 93/09239 ~
2 0 9 ~ ~~ e7 P~/US92/09769 Construction of Recombinant Plasmids WP-7A and pWP-19 The general overall strategy used to construct the prototype plasmid vectors, designated pWP-7A and pWP-19, respectively, is depicted in Figure 6. The XbaI
sites in plasmid psub201 were converted to EcoRl sites by ligating synthetic XbaI-EcoRl-XbaI adaptors as described by Srivastava et _al. (1989) Proc. Natl. Acad.
Sci. USA 86, 8078. The AAV coding region was removed following digestion with EcoRl, and the vector DNA
containing the two AAV-ITRs was isolated from preparative agarose gels (Seth [1984] Gene Anal. Tech.
1, 99), and treated with PolIk to generate blunt-ends.
Similarly, pBR322 DNA was cleaved with EcoRl and PvuII
and a 2066 by fragment containing the entire coding region of a gene for resistance to tetracycline (TcR) was also blunt ended with PolIk. These two fragments were ligated and used to transform competent E. coli HB101 cells by the standard methods described in Sambrook et al. (1989) to generate a plasmid, designated pWP-7A. Since blunt-end ligation of DNA fragments containing repaired EcoRl and PvuII ends regenerates an EcoRl site,~plasmid pWP-7A can be cleaved with EcoRl downstream from the TcR gene for cloning a gene or cassette of interest. The neon gene under the control of the herpesvirus thymidine kinase (TK) promoter was isolated from plasmid pSHL-172 (Tratschin _et _al. [1985]
Mol. Cell. Biol. 5, 3251) by partial digestion with PvuII, and blunt-end ligated with PolIk-treated pWP-7A
DNA. The resulting recombinant plasmid, designated pWP-SUBSTITUTE SHEET

~U~184~3 8A, is shown in Figure 6-A. Plasmid pWP-19 was constructed as follows. Plasmid pWP-8A was linearized with HindIII, which cleaves at the 5' end of the TcR
gene, and partially digested with XmnI to remove the TcR
gene. A plasmid pGBOR which contains a gene for resistance to ampicillin (ApR) and the bacteriophage lambda operator (OR1/OR2;lo) sequences (Samulski et al.
[1991] EMBO J. 10, 3941) was cleaved with EcoRl and XbaI
and the fragment containing the ~lo sequence was blunt-end ligated with Pollk-treated pWP-8A DNA described above. The resulting recombinant plasmid pWP-19 is shown in Figure 6-B.
The plasmid vectors pWP-7A and pWP-19 are useful for constructing recombinant AAV genomes because direct insertion of an insert of interest is possible in both, and the presence of the built-in neon gene in pWP-19 provides a strong selectable marker in human cells. The plasmid vector pWN-1 is particularly useful because it offers several features. In bacterial cells, these include: 1) The availability of a variety of cloning sites (EcoRl, BamHl, SacI, KpnI, XbaI, SalI, AccI, HincII, PstI, SphI and HindIII), including the NdeI
site; 2) AAV-ITRs which are well separated from the cloning sites; and 3) the use of TcR as well as ApR as selectable markers. In mammalian cells, following transfection in the presence of the AAV helper plasmid and Ad, the insert of interest, which is now flanked by the two AAV-ITRs in their proper orientation (see Figure 7), can be efficiently rescued from the TcR gene followed by DNA replication and packaging in the AAV
progeny virions.
SUBSTITUTE SHEET

WO 93/09239 2 0 9 g 4 ~ 3 PCT/US92/09?69 1 Whereas plasmid pWP-7A is useful for cloning inserts up to 2.5 kb in size at the unique EcoRl site, plasmid pWP-19 offers a built-in neon marker gene as well as a number of cloning sites such as BamHl, SacI
and KpnI, and insert up to 2.6 kb in size can be inserted between the two AAV-ITRs.

SUBST~'UTE SHEET

~U984-$~-1 RYSMDT.t7! Z
Construction of Recombinant Plasmid pWN-1 The strategy to construct the recombinant plasmid pWN-1 is shown in Figure 7. Briefly, psub201 plasmid DNA was digested to completion with XbaI and PvuII, and a 191-by XbaI-PvuII fragment containing the entire AAV-ITR sequence was isolated as described in Example 2.
Similarly, pBR322 plasmid DNA was cleaved with EcoRl and AvaI to isolate a 1425-by fragment that contains the TcR
gene but lacks the origin of DNA replication ("ori") sequence. This fragment was treated with PolIk to generate blunt ends. Blunt-ended EcoRl-AvaI fragment was mixed with a large excess of the XbaI-PvuII fragment containing the AAV-ITR, blunt-end ligated using T4 DNA
ligase, and then digested exhaustively with XbaI. This resulted in the production of the TcR gene flanked by a single AAV-ITR at each end but in the opposite orientation (see small arrows in boxed ITRs). This fragment was subsequently ligated at the unique XbaI
site in plasmid pGBOR described above, and TcR was used to select for the recombinant plasmid pWN-1.

SUBSTITUTE SHEET

WO 93/09239 ~ O 9 $ ~ ~ PCT/US92/09769 1 E~CAMPLE 4 Rescue and Replication of a C
DNA sequences flanked by the two AAV-ITRs can be rescued from the recombinant plasmids of Examples 2 and 3 following transfection in human cells in the presence of the AAV and Ad proteins as a prelude to successful packaging of these genes into mature AAV virions. The insert size between the two AAV-ITRs in plasmid pWP-8A
is similar to that of the wt AAV genome. The AAV-rep gene the parent plasmid psub201, as well as the x OR1/OR2 sequences from plasmid pGBOR were isolated and inserted in plasmid pWP-19 by the strategy shown in Figure 8-A. Two recombinant plasmids, pWP-21 and pWP-22, were generated which contain the AAV-rep gene in different orientations with respect to the neon gene.
These plasmids are depicted in Figure 8-A. Similarly, the insert size between the two AAV-ITRs in plasmid pWN-1 was increased by inserting the neon gene either at the NdeI site or at the PstI site to generate two recombinant plasmids, pWP-16 and pWP-17, respectively, which are shown in Figure 8-B.
Plasmids pWP-8A, pWP-21, pWP-22, pWP-16 and pWP-17 were either transfected alone, or co-transfected with the AAV helper plasmid (pAAV/Ad) separately, in Ad-infected human KB cells (Samulski _et _al. [1989] _J.
Virol. 63, 3822; Srivastava _et _al. [1989]; Srivastava [1990] Blood 76, 1997). Low M=. DNA samples isolated by the method described by Hirt (1967) J. Mol. Biol. _26, 365, were digested with DpnI and analyzed on Southern blots (Southern [1975] J. Mol. Biol. 98, 503) using a SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ ~ ~ PCT/US92/09769 neo-specific DNA probe as previously described (Samulski et al. 1989). The results are presented in Figure 9.
No rescue/replication of the recombinant neon gene from plasmid pWP-8A occurred in the absence of the pAAV/Ad helper plasmid (Lane 1); successful rescue and replication indeed occurred when the AAV-Rep proteins were supplied in trans (Lane 2), as detected by the presence of the characteristic monomeric and dimeric replicative intermediates of the recombinant AAV genome.
Similarly, rescue and replication occurred from plasmids pWP-21 (Lanes 3 and 4), and pWP-22 (Lanes 5 and 6) even in the absence of the helper plasmid because these plasmids contain the AAV-red gene in cis. Rescue and replication from plasmids pWP-16 and pWP-17 also occurred, but only in the presence of the AAV helper plasmid (Lanes 8 and 10).
Following rescue and replication, the neon gene could also be packaged into mature AAV progeny virions in presence of the AAV-Cap proteins. The recombinant AAV progeny virions were biologically active and infectious. For example, recombinant AAV-neo virions were used to transduce and stably integrate the neon gene in a variety of diploid and polyploid human cells.
The transduced neon gene was biologically active, as determined by gene expression analyses on Northern blots, as well as by ready isolation of clonal populations of human cells that were resistant to geneticin.

SI~BSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ ~ ~ ~ PGT/US92/09769 1 RXAMpT.R. S
Construction of an AAV-B19 Hybrid Parvovirus Clonincl and Expression Vector A hybrid vector containing the AAV-ITRs the B19p6 promoter and the neon gene as the heterologous gene was constructed as follows. The general strategy for the construction of this vector, designated pBl9-p6-Neor-AAV-ITR, is shown in Fig. 10.
A near full-length B19 DNA clone (for example pYT104v in which B19 is downstream of the bacteriophage SP6 promoter) was cleaved with SacI and DraI, and the small DNA fragment that contains the left ITR was discarded. Following blunt-end re-ligation of the larger fragment, the plasmid DNA was cleaved with EcoRI
and XbaI to isolate the 280 by fragment that contains the entire 5' non-coding region and the p6 promoter of B19. This fragment was cloned into the EcoRI-XbaI sites of pUCl9 to generate a plasmid pB19p6. This plasmid was cleaved with HincII which digests the B19 DNA downstream from the p6 promoter and also the pUCl9 DNA in the multiple cloning site. The bacterial Neor gene (Tratschin et al., 1985) was blunt-end ligated downstream from the B19p6 promoter between the two HincII sites to generate the plasmid pB19p6-Neor. This plasmid was digested with EcoRI and HindIII and the B19p6-Neo insert was isolated and ligated between the two AAV-ITRs of XbaI_digested psub201. The vector was packaged into mature AAV virions (vBl9-Neo) by cotransfection of adenovirus infected cells with pAAV/Ad, which contains the AAV-coding sequence and the 1 adenovirus type 5 terminal sequences (Samulski et al.
1989).

SUBSTITUTE SHEET

20884.83 1 RXAHIpT.R 6 Recombinant Parvovirus-Mediated Transfer of Bacterial Neor Gene in.Fiuman Hematopoietic Stem Cells Human hematopoietic stem cells were isolated from normal volunteer donors followed by sorting with monoclonal antibodies against the human CD34 and DR
antigens according to the method of Lu et al. (1987) J.
Immunol. 139, 1823 to produce a CD34''DR- cell Population. This cell population is known to contain several classes of primitive human hematopoietic progenitor cells including colony forming unit-blast cells (CFU-B1), high-proliferative potential colony forming cells (HPP-CFC), and cells responsible for initiating long-term hematopoiesis in vitro (LTBMIC).
Approximately 1 x 103 CD34'DR- cells isolated from two different donors were either mock-infected, or infected at varying moi with vTK-Neo or vHl9-Neo virions. (vTK-Neo is recombinant AAV virion containing the Neor gene under the control of the thymidine kinase (TK) promoter). Cells were incubated at 37°C for one week in the presence of the cytokines interleukin-3 (1 ng/ml), granulocyte macrophage colony stimulating factor (1 ng/ml), and a factor for c-kit ligand termed mast cell growth factor (50 ng/ml). 6418 was added at a final concentration of 250 ~.g/ml. The total number of viable cells was counted following one-week exposure to the drug. The concentration of 6418 was then increased to 500 ug/ml, and viable cell counts were obtained after two weeks for vTK-Neo-infected cells, and after one week for vBl9-Neo-infected cells. These data are shown in Fig. 11.
SUBSTITUTE SHEET

WO 93/09239 ~ ~ g ~ PCT/US92/09769 1 Exposure to vTK-Neo virions resulted in a nearly 10-fold increase in the 6418-resistant hematopoietic cell population compared with mock-infected cells, whereas the exposure to the vBl9-Neo virions resulted in approximately 4-fold increase at the highest moi of the virions compared with mock infected cells. These results demonstrate that the B19p6 promoter is active in cell populations enriched for HSC, albeit at a lower level compared with the TK promoter.

SUBSTITUTE SHEEfi WO 93/09239 ~ ~ ~ ~ ~ ~ PCT/US92/09769 Evaluation of Tissue-S ecificity of the B19 6 Promoter Non-erythroid cells were infected to determine 5whether the B19p6 promoter in the hybrid constructions had become indiscriminate or had maintained its erythroid specificity.
Human KB cells were either mock-infected or infected separately with equivalent moi of vTK-Neo and lOvBl9-Neo. At 48 hours post-infection cells were exposed to various concentration of 6418. Following a 14-day incubation period at 37°C, the approximate numbers of 6418-resistant colonies were enumerated. A colony is defined as a group of eight or more cells. These data l5are presented in Table 1, and demonstrate that under conditions of viral infection, the B19p6 promoter retains its erythroid-specificity.

Approximate Numbers of 6418-Resistant 20 Colonies in KB Cells Transduced with AAV-Neo Virions Recombinant l(i0 ~.g~'ml 400 virus 6418 wg/ml 6418 600 ug/ml 6418 1. None 10-20 0 0 2. vTK-Neo TMTC* 100-200 50-100 3. vBl9-Neo 10-20 0 0 *TMTC = Too many to count SUBSTITUTE SHEET

Construction of Parvovirus Vectors with an Erythroid-Specific Enhancer In order to further increase the tissue-specific expression directed by the B19p6 promoter, the DNasel-Hypersensitive Site-2 (HS-2) of the Locus Control Region (LCR), (Tuan et al. (1985) Proc. Natl. Acad. Sci. USA
82, 6384), an erythroid-specific enhancer, was inserted into the hybrid vectors of the present invention. As diagrammed in Fig. 12, the HS-2 gene is inserted upstream of the B19p6 promoter and luciferase gene to provide the vector vHS2/B19-Luc. Restriction sites used to facilitate vector construction are shown in Fig. 12.
These constructs were packaged into mature AAV virions.

SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ ~ ~ ~ PCT/US92/09769 1 1TY~M~T_Ti! O
Construction of Parvovirus Vectors Containing the Normal Human 13-Globin Gene In order to provide vectors for gene transfer in clinical cases of 13-thalassemia and sickle-cell anemia, two plasmid vectors that contain the normal human 13-globin gene were constructed and packaged into recombinant AAV virions. The pHS2/13-globin-Neo construct contains the a-globin promoter and the upstream HS-2 enhancer, along with the Neor gene under the control of the TK promoter, and the pHS2/B19-globin-Neo construct also contains the B19p6 promoter. These constructs are shown in Fig. 13.
The pHS2/Li-globin-Neo vector was constructed as follows. A plasmid (pWPl9) was constructed that contains the Neor gene under the TK promoter between the two AAV-ITRs. pWPl9 was linearized with Sacl. A SnaBI-PstI fragment containing the genomic clone of the human ~-globin gene was ligated, in reverse orientation, upstream of the TK promoter. The resulting plasmid was linearized by digestion with Kpnl, and a HindIII-XbaI
fragment containing the HS2 enhancer was ligated upstream of the f3-globin promoter.
The pHS2/B-19-globin-Neo vector was constructed as follows. First the HindIII-XbaI fragment containing the HS2 enhancer was cloned upstream of the 619p6 promoter in the plasmid pB19p6 by linearizing it with EcoRI. The HS2-B19p6 fragment was isolated by digesting this plasmid with PvuII and HincII. The PvuII-HincII
fragment was ligated to pWPl9 plasmid linearized with SacI. Second, the !3-globin coding region lacking the f3-SUBSTITUTE SHEET

1 globin promoter was excised by digesting the plasmid with NcoI and PstI. This NcoI-PstI fragment was ligated to the plasmid described above by linearizing it with KpnI.
Similarly, vectors containing the human a-globin gene in both orientations were constructed and packaged into recombinant AAV virions. A HinfI-PvuII fragment of the cloned a-globin gene (Liebhaber, 1980) was ligated downstream from the B19p6 promoter following linearization of the pB19p6 plasmid DNA. The resulting plasmid was linearized with Fspl, and ligated with the pWPl9 plasmid at the Sacl site prior to packaging into the AAV virions.

SUBSTITUTE SHEET

WO 93/09239 ~ PCT/US92/09769 AAV Tranduction of Human Cells with the MultidruQ Resistance Gene To assess the ability of AAV vectors to confer the multidrug resistance phenotype to drug sensitive human cells, drug sensitive human KB cells are transduced with the AAV vector containing an mdr gene under the control of the thymidine kinase promoter. The lU expression of the mdr phenotype is assessed by detection of P-glycoprotein with an antibody specific for this protein. Further, the ability of the transduced cells to reduce intracellular accumulation of certain anti-tumor drugs is evaluated by culturing transduced cells 15 in a series of tissue culture media containing step-wise concentration increases of the selected drug. An increase in viability of transduced cells relative to control (untransduced) cells is indicative of expression of the mdr gene .
To evaluate the ability of the AAV vectors of the present invention to confer cell-specific multidrug resistance, both erythroid and non-erythroid (e. g. drug sensitive KB) cells are transduced with the AAV vector containing an mdr gene under the control of the B19p6 25 promoter (AAV-B19p6-mdr). KB cells are transduced and assessed as described above. Since the B19p6 promoter directs erythroid cell-specific expression, the mdr gene is not expressed by transduction of KB cells with AAV-B19p6-mdr.
The ability of AAV-B19p6-mdr to confer erythroid cell-specific multidrug resistance is assessed as follows. Bone marrow is removed from a patient, SUBSTITUTE SHEET

~U9~$483 1 enriched to CD34~' cells and transduced with AAV-B19p6-mdr. Primary cultures are then established and maintained until focal colonies can be transferred to tissue culture trays. Outgrowth of such colonies permits the assessment of resistance to a range of anti-tumor drugs.
The AAV-B19p6-mdr vector is useful in cancer chemotherapy. For example, bone marrow is removed from the iliac creast of a cancer patient, and enriched for CD34~ cells. Enriched cells are transduced by AAV-B19p6-mdr and reinfused into the patient.
The B19p6 promoter can be substituted by other cell-specific promoter elements to permit expression of the multidrug resistance phenotype in other cell lineages.

SUBSTITUTE SHEET

WO 93/09239 ~ ~ ~ ~ ~ ~ ~ PCT/US92/09769 Cells of a human erythroleukemia cell line, K562 (ATCC CCL-243) were infected with vHS2/B19-globin-Neo (see Example 9 and Fig. 13) to assess the ability of the vector to provide expression of the B-globin gene in a host cell which exhibits no 13-globin gene expression.
K562 cells were either mock-infected or infected separately with equivalent moi of vHS2/B19-globin-Neo and vHS2/!3-globin-Neo. DNA of infected cells was isolated, digested with NcoI, and subjected to Southern blot analysis. Fig. 14 provides a Southern blot in which Lane 1 corresponds to DNA isolated from mock-infected cells, and Lane 2 corresponds to DNA isolated from cells infected with vHS2/a-globin-Neo. The blots were probed with a y-globin probe (left panel) and a f3-globin probe (right panel). The arrow denotes the transduced allele of the ti-globin gene in K562 cells infected with vHS2/13-globin-Neo. Similar results were obtained for K562 cells infected with vHS2/B19-globin-Neo.
The b-globin gene is present but not expressed in K562 cells. Northern blots of total RNA from the mock-infected and infected cells described above were probed with a neomycin probe (Fig. 15, left panel) and a f3-globin probe (Fig. 15, right panel). The center panel of Fig. 15 presents an ethidium-bromide stained gel.
Lane 1 in each panel represents RNA from mock-infected cells; Lanes 2 and 3 correspond to RNA from vHS2/!3-globin-Neo infected cells; Lanes 4 and 5 correspond to RNA from vHS2/B19-globin-Neo infected cells. Plus and minus signs indicate the presence or absence, SUBSTITUTE SHEET

WO 93/09239 , PCT/US92/09769 2p gg 48 3 respectively, of hemin, an inducer of y-globin which appeared to have no effect on b-globin expression. The Northern analysis indicates that the b-globin gene is expressed in K562 cells infected with vHS2/f~-globin-Neo and vHS2/B19-globin-Neo, but not in mock-infected cells.

3~
SUBSTITUTE SHEET

WO 93/09239 2 0 9 ~ 4 8 3 SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Srivastava, Arun (ii) TITLE OF INVENTION: SAFE VECTOR FOR GENE THERAPY
(iii) NUMBER OF SEQUENCES: 2 (iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Scully, Scott, Murphy Presser (B) STREET: 400 Garden City Plaza (C) CITY: Garden City (D) STATE: New York (E) COUNTRY: USA
(F) ZIP: 11530 (v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk (B) COMPUTER: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Release q.0, Version q.25 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: McNulty, William E.
(B) REGISTRATION NUMBER: 22,606 (C) REFERENCE/DOCKET NUMBER: 8361 (ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (516) 742-4343 (B) TELEFAX: (516)742-4366 (C) TELEX: 230 901 SANS UR
sussTrruTE sNE~

WO 93/09239 ~ PCT/US92/09769 (2) INFORMATION FOR SEQ ID NO:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 145 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:

GCCAAC'I'CCA TCACTAGGGG TTCCT 145 (2) INFORMATION FOR SEQ ID N0:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 225 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID N0:2:

SUBSTiT~TE SeiBET

Claims (46)

WHAT IS CLAIMED:
1. An expression vector for site-specific integration and cell-specific gene expression comprising two inverted terminal repeats of adeno-associated virus 2 and at least one cassette comprising a promoter capable of effecting cell-specific expression wherein said promoter is operably linked to a heterologous gene, and wherein said cassette resides between said inverted terminal repeats.
2. The vector of Claim 1 wherein each of said inverted terminal repeats comprises the nucleotides of SEQ ID NO:1.
3. The vector of Claim 1 wherein each of said inverted terminal repeats comprises nucleotides 1 to 125 of SEQ ID NO:1.
4. The vector of Claim 1 wherein said promoter is a B19 parvovirus promoter.
5. The vector of Claim 4 wherein said B19 parvovirus promoter is the p6 promoter.
6. The vector of Claim 4 wherein said B19 parvovirus promoter comprises the nucleotides of SEQ ID
NO:2.
7. The vector of Claim 1 wherein said heterologous gene encodes a biologically functional protein.
8. The vector of Claim 1 wherein said heterologous gene encodes a non-biologically functional protein.
9. The vector of Claim 1 wherein said heterologous gene encodes an antisense RNA.
10. The vector of Claim 1 wherein said heterologous gene is selected from the group consisting of a gene encoding .alpha.-globin, .beta.-globin, .gamma.-globin, granulocyte macrophage-colony stimulating factor (GM-CSF), tumor necrosis factor (TNF), any one of interleukins 1-11, neomycin resistance, luciferase, adenine phosphoribosyl transferase (APRT), retinoblastoma, insulin, mast cell growth factor, p53, adenosine deaminase.
11. The vector of Claim 1 wherein said heterologous gene encodes P-glycoprotein
12. The vector of Claim 9 wherein said antisense RNA
is complementary to a segment of the DNA or RNA encoding .alpha.-globin.
13. The vector of Claim 5 wherein said vector is AAV-B19-GM-CSF, AAV-B19-APRT, AAV-B19-neo r, AAV-B19-RB, AAV-B19-.beta.-globin, AAV-B19-.alpha.-globin, AAV-B19-.gamma.-globin.
14. The vector of Claim 5 wherein said vector is AAB-B19-mdr.
15. The vector of any one of Claims 1-14 wherein said vector is contained in a host cell.
16. The vector of any one of Claims 1-12 wherein said vector is contained in a virion.
17. The vector of Claim 16 wherein said vector is contained in a host cell.
18. The vector of Claim 15 or 17 wherein said host cell is a hematopoietic stem or progenitor cell.
19. Use of the vector of Claim 1 for gene therapy.
20. The use of Claim 19 wherein said use involves transfection with said vector or by infection with virions containing said vector.
21. The use of Claim 19 wherein said gene therapy comprises treatment of sickle-cell anemia or diabetes.
22. The use of Claim 19 wherein said gene therapy comprises treatment of thalassemia.
23. The use of Claim 22 wherein said vector is AAV-B19-.alpha.-globin or AAV-B19-.beta.-globin.
24. The use of Claim 19 wherein said gene therapy comprises treatment of hematopoietic diseases.
25. The use of Claim 24 wherein said vector is AAV-B19-GM-CSF.
26. The use of Claim 24 wherein said gene therapy comprises treatment of cancer.
27. The use of Claim 26 wherein said vector is AAV-B19-RB.
28. The use of Claim 19 wherein said gene therapy comprises treatment of hemoglobinopathies.
29. The use of Claim 28 wherein said vector is AAV-B19-.beta.-globin, AAV-B19-.alpha.-globin or AAV-B19-.gamma.-globin.
30. Use of the vector of Claim 1 for delivering a pharmaceutical product.
31. The use of Claim 30 wherein said use involves transfection or by infection with virions containing said vector.
32. The use of Claim 30 wherein said product is .gamma.-globin, insulin, macrophage colony stimulating factor, granulocyte colony stimulating factor, erythropoietin, tumor necrosis factor, mast cell growth factor, any of interleukins 1-11, p53, adenosine deaminase or an antisense RNA molecule.
33. The use of Claim 30 wherein said product is granulocyte macrophage-colony stimulating factor.
34. The use of Claim 33 wherein said vector is AAB-B19-GM-CSF.
35. The use of Claim 30 wherein said product is .alpha.-globin.
36. The use of Claim 35 wherein said vector is AAV-B19-.alpha.-globin.
37. The use of Claim 30 wherein said product is .beta.-globin.
38. The use of Claim 37 wherein said vector is AAV-B19-.beta.-globin.
39. The use of Claim 30 wherein said product is retinoblastoma.
40. The use of Claim 39 wherein said vector is AAV-B19-RB.
41. Use of the vector of Claim 11 for conferring cell-specific multidrug resistance.
42. Use of the vector of Claim 14 for conferring multidrug resistance to erythroid cells.
43. The use of Claim 41 or 42 wherein said use involves transfection with said vector or by infection with virions containing said vector.
44. An expression vector comprising two inverted terminal repeats of adeno-associated virus 2 and at least one cassette comprising a promoter capable of effecting cell-specific expression wherein said promoter is operably linked to a heterologous gene, and wherein said cassette resides between said inverted terminal repeats.
45. Use of the vector of Claim 44 for gene therapy.
46. Use of the vector of Claim 44 for delivering a pharmaceutical product.
CA002098483A 1991-11-08 1992-11-06 Safe adeno-associated virus 2 vector for gene therapy Expired - Fee Related CA2098483C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US789,917 1991-11-08
US07/789,917 US5252479A (en) 1991-11-08 1991-11-08 Safe vector for gene therapy
PCT/US1992/009769 WO1993009239A1 (en) 1991-11-08 1992-11-06 Adeno-associated virus-2 basal vectors

Publications (2)

Publication Number Publication Date
CA2098483A1 CA2098483A1 (en) 1993-05-09
CA2098483C true CA2098483C (en) 2000-03-14

Family

ID=25149101

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002098483A Expired - Fee Related CA2098483C (en) 1991-11-08 1992-11-06 Safe adeno-associated virus 2 vector for gene therapy

Country Status (13)

Country Link
US (1) US5252479A (en)
EP (1) EP0566732B1 (en)
JP (2) JPH06504680A (en)
KR (1) KR930703453A (en)
AT (1) ATE236263T1 (en)
AU (1) AU657829B2 (en)
CA (1) CA2098483C (en)
DE (1) DE69232983T2 (en)
DK (1) DK0566732T3 (en)
ES (1) ES2192555T3 (en)
IL (1) IL103677A0 (en)
PT (1) PT101040B (en)
WO (1) WO1993009239A1 (en)

Families Citing this family (516)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6605712B1 (en) * 1990-12-20 2003-08-12 Arch Development Corporation Gene transcription and ionizing radiation: methods and compositions
AU663702B2 (en) 1991-03-06 1995-10-19 Board Of Regents, The University Of Texas System Methods and compositions for the selective inhibition of gene expression
US5747469A (en) * 1991-03-06 1998-05-05 Board Of Regents, The University Of Texas System Methods and compositions comprising DNA damaging agents and p53
US6410010B1 (en) * 1992-10-13 2002-06-25 Board Of Regents, The University Of Texas System Recombinant P53 adenovirus compositions
US6261834B1 (en) * 1991-11-08 2001-07-17 Research Corporation Technologies, Inc. Vector for gene therapy
US5587308A (en) * 1992-06-02 1996-12-24 The United States Of America As Represented By The Department Of Health & Human Services Modified adeno-associated virus vector capable of expression from a novel promoter
US6268213B1 (en) * 1992-06-03 2001-07-31 Richard Jude Samulski Adeno-associated virus vector and cis-acting regulatory and promoter elements capable of expressing at least one gene and method of using same for gene therapy
US6348352B1 (en) 1992-09-18 2002-02-19 Canji, Inc. Methods for selectively transducing pathologic mammalian cells using a tumor suppressor gene
US5658565A (en) * 1994-06-24 1997-08-19 University Of Pittsburgh Of The Commonwealth System Of Higher Education Inducible nitric oxide synthase gene for treatment of disease
US5830461A (en) * 1992-11-25 1998-11-03 University Of Pittsburgh Of The Commonwealth System Of Higher Education Methods for promoting wound healing and treating transplant-associated vasculopathy
US6340674B1 (en) 1993-03-26 2002-01-22 Thomas Jefferson University Method of inhibiting the proliferation and causing the differentiation of cells with IGF-1 receptor antisense oligonucleotides
DE69432409T2 (en) 1993-04-30 2003-12-24 Wellstat Biologics Corp Use of the NDV to manufacture a drug for cancer treatment
US6686200B1 (en) * 1993-08-31 2004-02-03 Uab Research Foundation Methods and compositions for the large scale production of recombinant adeno-associated virus
WO1997003703A1 (en) * 1995-07-21 1997-02-06 Rhone-Poulenc Rorer Pharmaceuticals Inc. Adeno-associated viral liposomes and their use in transfecting dendritic cells to stimulate specific immunity
US6652850B1 (en) 1993-09-13 2003-11-25 Aventis Pharmaceuticals Inc. Adeno-associated viral liposomes and their use in transfecting dendritic cells to stimulate specific immunity
US5834441A (en) * 1993-09-13 1998-11-10 Rhone-Poulenc Rorer Pharmaceuticals Inc. Adeno-associated viral (AAV) liposomes and methods related thereto
US5693531A (en) * 1993-11-24 1997-12-02 The United States Of America As Represented By The Department Of Health And Human Services Vector systems for the generation of adeno-associated virus particles
FR2716893B1 (en) * 1994-03-03 1996-04-12 Rhone Poulenc Rorer Sa Recombinant viruses, their preparation and their therapeutic use.
IL113052A0 (en) 1994-03-23 1995-06-29 Rhone Poulenc Rorer Sa Recombinant viruses, their preparation and their use in gene therapy
DE4411402A1 (en) * 1994-03-31 1995-10-05 Juergen Schrader DNA expression vectors for use in gene therapy treatment of vascular diseases
WO1995027494A1 (en) * 1994-04-11 1995-10-19 Sloan-Kettering Institute For Cancer Research DEFECTIVE HERPES AND DEFECTIVE ADENO-ASSOCIATED VIRUS VECTORS WITH p53 FOR THE TREATMENT OF CANCER
DE69535703T2 (en) * 1994-04-13 2009-02-19 The Rockefeller University AAV-mediated delivery of DNA to cells of the nervous system
WO1995032225A1 (en) * 1994-05-23 1995-11-30 The Salk Institute For Biological Studies Method for site-specific integration of nucleic acids and related products
US6814962B1 (en) * 1994-06-02 2004-11-09 Aventis Pharma S.A. Recombinant viruses and their use for treatment of atherosclerosis and other forms of coronary artery disease and method, reagent, and kit for evaluating susceptibility to same
US20020159979A1 (en) 1994-06-06 2002-10-31 Children's Hospital, Inc. Adeno-associated virus materials and methods
US5658785A (en) * 1994-06-06 1997-08-19 Children's Hospital, Inc. Adeno-associated virus materials and methods
US6204059B1 (en) * 1994-06-30 2001-03-20 University Of Pittsburgh AAV capsid vehicles for molecular transfer
MX9701075A (en) 1994-08-12 1998-03-31 Myriad Genetics Inc IN VIVO MUTATIONS AND POLYMORPHISMS IN THE 17q-LINKED BREAST AND OVARIAN CANCER SUSCEPTIBILITY GENE.
ATE198623T1 (en) 1994-08-12 2001-01-15 Myriad Genetics Inc METHOD FOR DETECTING PREDISPOSITION OF OVARIAL AND BREAST CANCER
US5871986A (en) 1994-09-23 1999-02-16 The General Hospital Corporation Use of a baculovirus to express and exogenous gene in a mammalian cell
US5652225A (en) * 1994-10-04 1997-07-29 St. Elizabeth's Medical Center Of Boston, Inc. Methods and products for nucleic acid delivery
DK0787200T3 (en) * 1994-10-28 2005-08-15 Univ Pennsylvania Improved adenovirus and methods for its use
US5856152A (en) * 1994-10-28 1999-01-05 The Trustees Of The University Of Pennsylvania Hybrid adenovirus-AAV vector and methods of use therefor
US5872005A (en) * 1994-11-03 1999-02-16 Cell Genesys Inc. Packaging cell lines for adeno-associated viral vectors
US5714170A (en) 1994-11-16 1998-02-03 Thomas Jefferson University Method of inducing resistance to tumor growth
US5998205A (en) 1994-11-28 1999-12-07 Genetic Therapy, Inc. Vectors for tissue-specific replication
US6638762B1 (en) 1994-11-28 2003-10-28 Genetic Therapy, Inc. Tissue-vectors specific replication and gene expression
FR2731710B1 (en) * 1995-03-14 1997-04-30 Rhone Poulenc Rorer Sa RECOMBINANT VIRUSES EXPRESSING LECITHIN CHOLESTEROL ACYLTRANSFERASE AND USES IN GENE THERAPY
AU4954896A (en) * 1995-03-16 1996-10-02 Hisamitsu Pharmaceutical Co., Inc. Novel cell strains
FR2732357B1 (en) * 1995-03-31 1997-04-30 Rhone Poulenc Rorer Sa VIRAL VECTORS AND USE FOR THE TREATMENT OF HYPERPROLIFERATIVE DISORDERS, ESPECIALLY RESTENOSIS
DE19513152A1 (en) * 1995-04-07 1996-10-10 Bundesrep Deutschland Use of an "immunodeficiency virus suppressing lymphokine (ISL)" to inhibit virus multiplication, in particular of retroviruses
US7745416B2 (en) * 1995-04-11 2010-06-29 The Regents Of The University Of California Method for in vivo regulation of cardiac muscle contractility
US6638264B1 (en) * 1995-05-16 2003-10-28 Biostratum Incorporation Perfusion apparatus and methods for pharmaceutical delivery
AU5658196A (en) * 1995-05-16 1996-11-29 Pirkko Heikkila Method for pharmaceutical delivery
WO1996037626A1 (en) * 1995-05-22 1996-11-28 Chiron Corporation Position-specific integration of vector constructs into eukaryotic genomes mediated by a chimeric integrase protein
US6281010B1 (en) 1995-06-05 2001-08-28 The Trustees Of The University Of Pennsylvania Adenovirus gene therapy vehicle and cell line
US5756283A (en) * 1995-06-05 1998-05-26 The Trustees Of The University Of Pennsylvania Method for improved production of recombinant adeno-associated viruses for gene therapy
US6187757B1 (en) * 1995-06-07 2001-02-13 Ariad Pharmaceuticals, Inc. Regulation of biological events using novel compounds
US6110456A (en) * 1995-06-07 2000-08-29 Yale University Oral delivery or adeno-associated viral vectors
US5990388A (en) * 1995-06-07 1999-11-23 Research Corporation Technologies, Inc. Resistance to viruses and viroids in transgenic plants and animals expressing dsRNA-binding protein
US20020068049A1 (en) * 1998-09-10 2002-06-06 Henderson Daniel R. Tissue specific adenoviral vectors
US5770720A (en) * 1995-08-30 1998-06-23 Barnes-Jewish Hospital Ubiquitin conjugating enzymes having transcriptional repressor activity
US5795872A (en) * 1995-09-19 1998-08-18 Pharmadigm, Inc. DNA construct for immunization
US6162796A (en) * 1995-09-27 2000-12-19 The Rockefeller University Method for transferring genes to the heart using AAV vectors
WO1997018241A1 (en) * 1995-11-14 1997-05-22 Thomas Jefferson University Inducing resistance to tumor growth with soluble igf-1 receptor
ATE325872T1 (en) * 1995-11-30 2006-06-15 Univ Texas METHODS AND COMPOSITIONS FOR TREATING CANCER
DE69625678T3 (en) 1995-12-18 2006-11-09 The University Of Utah Research Foundation, Salt Lake City Chromosome 13 associated breast cancer susceptibility gene BRCA2
US5837492A (en) 1995-12-18 1998-11-17 Myriad Genetics, Inc. Chromosome 13-linked breast cancer susceptibility gene
NZ330744A (en) 1995-12-22 1999-02-25 Univ Utah Res Found A long qt syndrome gene which encodes kvlqt1 and its association with mink
US5846528A (en) * 1996-01-18 1998-12-08 Avigen, Inc. Treating anemia using recombinant adeno-associated virus virions comprising an EPO DNA sequence
US5962313A (en) 1996-01-18 1999-10-05 Avigen, Inc. Adeno-associated virus vectors comprising a gene encoding a lyosomal enzyme
US5858351A (en) * 1996-01-18 1999-01-12 Avigen, Inc. Methods for delivering DNA to muscle cells using recombinant adeno-associated virus vectors
DE19608751B4 (en) * 1996-03-06 2006-05-18 Medigene Ag Use of an adeno-associated virus vector to increase the immunogenicity of cells
US5952221A (en) * 1996-03-06 1999-09-14 Avigen, Inc. Adeno-associated virus vectors comprising a first and second nucleic acid sequence
GB9605124D0 (en) 1996-03-11 1996-05-08 Royal Free Hosp School Med Method of treating muscular disorders
US6723531B2 (en) 1996-04-05 2004-04-20 The Salk Institute For Biological Studies Method for modulating expression of exogenous genes in mammalian systems, and products related thereto
AU3130597A (en) * 1996-05-16 1997-12-05 Duke University Tristetraprolin
US5780447A (en) * 1996-06-14 1998-07-14 St. Jude Children's Research Hospital Recombinant adeno-associated viral vectors
US6534314B1 (en) 1996-06-14 2003-03-18 Massachusetts Institute Of Technology Methods and compositions for transforming cells
US5928914A (en) * 1996-06-14 1999-07-27 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva University Methods and compositions for transforming cells
EP0979101B1 (en) 1996-07-03 2010-10-27 Merial, Inc. Recombinant canine adenovirus 2 (cav2) containing exogenous dna
US20020037867A1 (en) * 1999-02-26 2002-03-28 James M. Wilson Method for recombinant adeno-associated virus-directed gene therapy
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
JP2001500376A (en) 1996-09-06 2001-01-16 カイロン コーポレイション Methods and compositions for liver-specific delivery of therapeutic molecules using recombinant AAV vectors
US6281009B1 (en) 1996-09-11 2001-08-28 The General Hospital Corporation Use of a non-mammalian DNA virus to express an exogenous gene in a mammalian cell
US6544523B1 (en) 1996-11-13 2003-04-08 Chiron Corporation Mutant forms of Fas ligand and uses thereof
WO1998024924A1 (en) * 1996-12-05 1998-06-11 Introgene B.V. Genetic modification of primate hemopoietic repopulating stem cells
US7008776B1 (en) 1996-12-06 2006-03-07 Aventis Pharmaceuticals Inc. Compositions and methods for effecting the levels of high density lipoprotein (HDL) cholesterol and apolipoprotein AI very low density lipoprotein (VLDL) cholesterol and low density lipoprotein (LDL) cholesterol
GB9701425D0 (en) * 1997-01-24 1997-03-12 Bioinvent Int Ab A method for in vitro molecular evolution of protein function
US6218597B1 (en) 1997-04-03 2001-04-17 University Technology Corporation Transgenic model and treatment for heart disease
AU753781B2 (en) 1997-04-28 2002-10-31 Aventis Pharma S.A. Adenovirus-mediated intratumoral delivery of an angiogenesis antagonist for the treatment of tumors
US6541036B1 (en) 1997-05-29 2003-04-01 Thomas Jefferson University Treatment of tumors with oligonucleotides directed to insulin-like growth factor-I receptors (IGF-IR)
US6156303A (en) * 1997-06-11 2000-12-05 University Of Washington Adeno-associated virus (AAV) isolates and AAV vectors derived therefrom
US6635623B1 (en) 1997-06-13 2003-10-21 Baylor College Of Medicine Lipoproteins as nucleic acid vectors
US20050096288A1 (en) * 1997-06-13 2005-05-05 Aragene, Inc. Lipoproteins as nucleic acid vectors
GB9712512D0 (en) 1997-06-16 1997-08-20 Bioinvent Int Ab A method for in vitro molecular evolution of protein function
US6300488B1 (en) 1997-07-10 2001-10-09 The Salk Institute For Biological Studies Modified lepidopteran receptors and hybrid multifunctional proteins for use in transcription and regulation of transgene expression
US6251677B1 (en) 1997-08-25 2001-06-26 The Trustees Of The University Of Pennsylvania Hybrid adenovirus-AAV virus and methods of use thereof
EP1027451A1 (en) * 1997-10-08 2000-08-16 Advanced Research & Technology Institute Chimeric parvovirus-based recombinant vector system that specifically targets the erythroid lineage
US6346415B1 (en) * 1997-10-21 2002-02-12 Targeted Genetics Corporation Transcriptionally-activated AAV inverted terminal repeats (ITRS) for use with recombinant AAV vectors
EP2147681A1 (en) 1997-10-29 2010-01-27 Genzyme Corporation Compositions and methods for treating lysosomal storage disease
CN1263854C (en) 1997-11-06 2006-07-12 启龙股份公司 Neisserial antigens
AU759707B2 (en) 1998-01-08 2003-04-17 Aventis Pharmaceuticals Products Inc. A transgenic rabbit that expresses a functional human lipoprotein (A)
US6780609B1 (en) 1998-10-23 2004-08-24 Genome Therapeutics Corporation High bone mass gene of 1.1q13.3
SG152917A1 (en) 1998-01-14 2009-06-29 Chiron Srl Neisseria meningitidis antigens
US6984635B1 (en) 1998-02-13 2006-01-10 Board Of Trustees Of The Leland Stanford Jr. University Dimerizing agents, their production and use
EP2261339B1 (en) 1998-05-01 2017-03-22 GlaxoSmithKline Biologicals SA Neisseria meningitidis antigens and compositions
US6225456B1 (en) 1998-05-07 2001-05-01 University Technololy Corporation Ras suppressor SUR-5
US6333318B1 (en) * 1998-05-14 2001-12-25 The Salk Institute For Biological Studies Formulations useful for modulating expression of exogenous genes in mammalian systems, and products related thereto
US6506889B1 (en) 1998-05-19 2003-01-14 University Technology Corporation Ras suppressor SUR-8 and related compositions and methods
US7153655B2 (en) * 1998-06-16 2006-12-26 Alligator Bioscience Ab Method for in vitro molecular evolution of protein function involving the use of exonuclease enzyme and two populations of parent polynucleotide sequence
US6135976A (en) 1998-09-25 2000-10-24 Ekos Corporation Method, device and kit for performing gene therapy
US6743906B1 (en) 1998-10-02 2004-06-01 Board Of Regents, The University Of Texas PPP2R1B is a tumor suppressor
US6221349B1 (en) 1998-10-20 2001-04-24 Avigen, Inc. Adeno-associated vectors for expression of factor VIII by target cells
US6200560B1 (en) * 1998-10-20 2001-03-13 Avigen, Inc. Adeno-associated virus vectors for expression of factor VIII by target cells
US7129043B1 (en) 1998-10-22 2006-10-31 Duke University Methods of screening for risk of proliferative disease and methods for the treatment of proliferative disease
US6759237B1 (en) 1998-11-05 2004-07-06 The Trustees Of The University Of Pennsylvania Adeno-associated virus serotype 1 nucleic acid sequences, vectors and host cells containing same
US6855549B1 (en) 1998-11-23 2005-02-15 The University Of Iowa Research Foundation Methods and compositions for increasing the infectivity of gene transfer vectors
US6441156B1 (en) * 1998-12-30 2002-08-27 The United States Of America As Represented By The Department Of Health And Human Services Calcium channel compositions and methods of use thereof
US6387368B1 (en) 1999-02-08 2002-05-14 The Trustees Of The University Of Pennsylvania Hybrid adenovirus-AAV virus and methods of use thereof
US6495376B1 (en) 1999-02-18 2002-12-17 Beth Israel Deaconess Medical Center Methods and compositions for regulating protein-protein interactions
US7794703B1 (en) * 1999-02-19 2010-09-14 Hai Xing Chen Method for production and delivery of a protein in vivo
US20030215423A1 (en) * 1999-04-01 2003-11-20 Merck & Co., Inc. Gene therapy for obesity
US20080145347A1 (en) * 1999-04-29 2008-06-19 Steiner Mitchell S p-Hyde sequences in the Rat
WO2000066741A2 (en) 1999-04-30 2000-11-09 Chiron S.P.A. Conserved neisserial antigens
GB9911683D0 (en) 1999-05-19 1999-07-21 Chiron Spa Antigenic peptides
US6165754A (en) * 1999-06-08 2000-12-26 Cornell Research Foundation, Inc. Method of expressing an exogenous nucleic acid
US7504253B2 (en) * 1999-06-11 2009-03-17 The Burnham Institute For Medical Research Nucleic acid encoding proteins involved in protein degradation, products and methods related thereof
GB9916529D0 (en) 1999-07-14 1999-09-15 Chiron Spa Antigenic peptides
CA2382045A1 (en) * 1999-08-20 2001-03-01 Board Of Regents, The University Of Texas System Hdac4 and hdac5 in the regulation of cardiac gene expression
US7057015B1 (en) 1999-10-20 2006-06-06 The Salk Institute For Biological Studies Hormone receptor functional dimers and methods of their use
BR0015137A (en) 1999-10-29 2003-03-25 Chiron Spa Neisserial antigenic peptides
WO2001036623A2 (en) * 1999-11-05 2001-05-25 Avigen, Inc. Ecdysone-inducible adeno-associated virus expression vectors
US6582692B1 (en) * 1999-11-17 2003-06-24 Avigen, Inc. Recombinant adeno-associated virus virions for the treatment of lysosomal disorders
WO2001036640A2 (en) 1999-11-18 2001-05-25 Chiron Corporation Human fgf-21 gene and gene expression products
US6241710B1 (en) 1999-12-20 2001-06-05 Tricardia Llc Hypodermic needle with weeping tip and method of use
RU2279889C2 (en) 2000-01-17 2006-07-20 Чирон С.Р.Л. OUTER MEMBRANE VESICLE VACCINE (OMV) CONTAINING PROTEINS OF SEROGROUP B N.Meningitis OUTER MEMBRANE
EP1854476A3 (en) 2000-02-09 2008-05-07 Bas Medical, Inc. Use of relaxin to treat diseases related to vasoconstriction
EP1259626B1 (en) * 2000-02-25 2007-10-31 Ludwig Institute For Cancer Research Materials and methods involving hybrid vascular endothelial growth factor dnas and proteins and screening methods for modulators
WO2001066595A2 (en) 2000-03-08 2001-09-13 Chiron Corporation Human fgf-23 gene and gene expression products
GB0018307D0 (en) 2000-07-26 2000-09-13 Aventis Pharm Prod Inc Polypeptides
EP1950297A2 (en) 2000-05-31 2008-07-30 Novartis Vaccines and Diagnostics, Inc. Compositions and methods for treating neoplastic disease using chemotherapy and radiation sensitizers
US7198924B2 (en) 2000-12-11 2007-04-03 Invitrogen Corporation Methods and compositions for synthesis of nucleic acid molecules using multiple recognition sites
NZ594877A (en) 2000-10-27 2012-07-27 Novartis Vaccines & Diagnostic Nucleic acids and proteins from streptococcus groups A & B
JP2004536555A (en) 2000-11-03 2004-12-09 ダナ ファーバー キャンサー インスティテュート Methods and compositions for diagnosing cancer susceptibility and defective DNA repair mechanisms and treatments
US20030188326A1 (en) 2000-11-03 2003-10-02 Dana Farber Cancer Institute Methods and compositions for the diagnosis of cancer susceptibilities and defective DNA repair mechanisms and treatment thereof
US6958213B2 (en) 2000-12-12 2005-10-25 Alligator Bioscience Ab Method for in vitro molecular evolution of protein function
US20020086292A1 (en) 2000-12-22 2002-07-04 Shigeaki Harayama Synthesis of hybrid polynucleotide molecules using single-stranded polynucleotide molecules
MXPA03005386A (en) 2000-12-28 2004-04-20 Wyeth Corp Recombinant protective protein from $i(streptococcus pneumoniae).
US6613534B2 (en) 2001-03-20 2003-09-02 Wake Forest University Health Sciences MAP-2 as a determinant of metastatic potential
GB0107658D0 (en) 2001-03-27 2001-05-16 Chiron Spa Streptococcus pneumoniae
GB0107661D0 (en) 2001-03-27 2001-05-16 Chiron Spa Staphylococcus aureus
ATE541937T1 (en) * 2001-04-06 2012-02-15 Univ Chicago CHEMOTHERAPEUTIC INTRODUCTION OF EGR-1 PROMOTER ACTIVITY IN GENE THERAPY
US20040242523A1 (en) * 2003-03-06 2004-12-02 Ana-Farber Cancer Institue And The Univiersity Of Chicago Chemo-inducible cancer gene therapy
WO2002081639A2 (en) 2001-04-06 2002-10-17 Georgetown University Gene brcc2 and diagnostic and therapeutic uses thereof
WO2002081641A2 (en) 2001-04-06 2002-10-17 Georgetown University Gene scc-112 and diagnostic and therapeutic uses thereof
WO2002081642A2 (en) 2001-04-06 2002-10-17 Georgetown University Gene brcc-3 and diagnostic and therapeutic uses thereof
US8034791B2 (en) 2001-04-06 2011-10-11 The University Of Chicago Activation of Egr-1 promoter by DNA damaging chemotherapeutics
US8067156B2 (en) 2001-05-31 2011-11-29 The Rockefeller University Method for generating replication defective viral vectors that are helper free
AUPR546801A0 (en) 2001-06-05 2001-06-28 Commonwealth Scientific And Industrial Research Organisation Recombinant antibodies
US7449562B2 (en) * 2001-06-29 2008-11-11 Novartis Ag PERV screening method and use thereof
MX339524B (en) 2001-10-11 2016-05-30 Wyeth Corp Novel immunogenic compositions for the prevention and treatment of meningococcal disease.
DE60228758D1 (en) 2001-12-12 2008-10-16 Novartis Vaccines & Diagnostic IMMUNIZATION AGAINST CHLAMYDIA TRACHEOMATIS
WO2003078453A1 (en) 2002-03-15 2003-09-25 Wyeth Holdings Corporation Mutants of the p4 protein of nontypable haemophilus influenzae with reduced enzymatic activity
US7384738B2 (en) * 2002-03-28 2008-06-10 Bremel Robert D Retrovirus-based genomic screening
US7244565B2 (en) 2002-04-10 2007-07-17 Georgetown University Gene shinc-3 and diagnostic and therapeutic uses thereof
WO2003087367A2 (en) 2002-04-18 2003-10-23 Lynkeus Biotech Gmbh Means and methods for the specific inhibition of genes in cells and tissue of the cns and/or eye
JP4372679B2 (en) * 2002-05-17 2009-11-25 アリゲーター・バイオサイエンス・アーベー In vitro molecular evolution of protein function
EP2316922B1 (en) 2002-05-24 2013-05-22 Merck Sharp & Dohme Corp. Neutralizing human anti-IGFR antibody
US7148342B2 (en) 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
US7785608B2 (en) 2002-08-30 2010-08-31 Wyeth Holdings Corporation Immunogenic compositions for the prevention and treatment of meningococcal disease
US20040152112A1 (en) 2002-11-13 2004-08-05 Thomas Jefferson University Compositions and methods for cancer diagnosis and therapy
WO2004050683A2 (en) 2002-12-02 2004-06-17 Abgenix, Inc. Antibodies directed to tumor necrosis factor and uses thereof
US20050048041A1 (en) * 2003-01-13 2005-03-03 Rao Mahendra S. Persistent expression of candidate molecule in proliferating stem and progenitor cells for delivery of therapeutic products
EP1604010B1 (en) 2003-01-16 2010-08-11 The Trustees of The University of Pennsylvania COMPOSITIONS AND METHODS FOR siRNA INHIBITION OF ICAM-1
GB0308198D0 (en) 2003-04-09 2003-05-14 Chiron Srl ADP-ribosylating bacterial toxin
US20050028229A1 (en) * 2003-04-14 2005-02-03 Mitchell Weiss Alpha-hemoglobin stabilizing protein transgenic mouse and methods of use thereof
EP1629008A1 (en) * 2003-05-21 2006-03-01 Ares Trading S.A. Tnf-like secreted protein
JP2007505603A (en) * 2003-09-12 2007-03-15 ヴァーテックス ファーマシューティカルズ、 インコーポレイテッド Animal models for protease activity and liver injury
US20060078558A1 (en) * 2003-11-12 2006-04-13 Whitsett Jeffrey A Diagnosis, prognosis and treatment of pulmonary diseases
JP2007512838A (en) 2003-12-01 2007-05-24 インヴィトロジェン コーポレーション Nucleic acid molecules containing recombination sites and methods of use thereof
WO2005067708A2 (en) 2004-01-14 2005-07-28 Daniolabs Limited Zebrafish model for autoimmune diseases
US7432057B2 (en) 2004-01-30 2008-10-07 Michigan State University Genetic test for PSE-susceptible turkeys
EP2295604B1 (en) 2004-02-09 2015-04-08 Thomas Jefferson University Diagnosis and treatment of cancers with microRNA located in or near cancer-associated chromosomal features
DE602005027673D1 (en) 2004-03-05 2011-06-09 Vegenics Pty Ltd MATERIALS AND METHODS FOR GROWTH FACTORY BONDING CONSTRUCTS
EP2365077B1 (en) * 2004-03-12 2013-05-08 Alnylam Pharmaceuticals, Inc. iRNA agents targeting VEGF
US7582442B2 (en) 2004-03-16 2009-09-01 The Regents Of The University Of Michigan Methods and compositions for using aleveolar macrophage phospholipase A2
US7319015B2 (en) * 2004-03-16 2008-01-15 The Regents Of The University Of Michigan Methods and compositions for using alveolar macrophage phospholipase A2
CN104292321A (en) 2004-03-29 2015-01-21 株式会社嘉尔药物 Novel modified galectin 9 protein and use thereof
US7604798B2 (en) 2004-07-15 2009-10-20 Northwestern University Methods and compositions for importing nucleic acids into cell nuclei
WO2006014798A2 (en) * 2004-07-27 2006-02-09 Mount Sinai School Of Medicine Methods and compositions for using sax2
ATE484280T1 (en) * 2004-08-25 2010-10-15 Univ Chicago USE OF THE COMBINATION OF TEMOZOLOMIDE AND TNF-ALPHA FOR THE TREATMENT OF GLIOBLASTOMA
FI20050753A (en) 2004-09-03 2006-03-04 Licentia Oy New peptides
GB0423126D0 (en) * 2004-10-18 2004-11-17 Ares Trading Sa Protein
JP2008520583A (en) * 2004-11-15 2008-06-19 マウント シナイ スクール オブ メディスン オブ ニューヨーク ユニバーシティー Compositions and methods for modifying WNT autocrine signaling
GB0426960D0 (en) 2004-12-08 2005-01-12 Ares Trading Sa TGR-3 like protein receptor
US7531523B2 (en) * 2005-02-17 2009-05-12 Vertex Pharmaceuticals Incorporated Sodium channel protein type III alpha-subunit splice variant
GB0504767D0 (en) * 2005-03-08 2005-04-13 Ares Trading Sa Lipocalin protein
US20060216315A1 (en) * 2005-03-16 2006-09-28 Yoo Tai J Cockroach allergen gene expression and delivery systems and uses
US20100028312A1 (en) * 2005-03-24 2010-02-04 Caritas St. Elizabeth Medical Center of Boston Inc Stably transformed bone marrow-derived cells and uses thereof
EP3782655A1 (en) 2005-05-17 2021-02-24 Amicus Therapeutics, Inc. A method for the treatment of pompe disease using 1-deoxynojirimycin and derivatives
WO2007008252A1 (en) * 2005-07-12 2007-01-18 Temple University - Of The Commonwealth Systems Of Higher Education Genetic and epigenetic alterations in the diagnosis and treatment of cancer
WO2007016643A2 (en) * 2005-08-01 2007-02-08 Mount Sinai School Of Medicine Of New York University A method for extending longevity using npc1l1 antagonists
GB2429013C (en) 2005-08-11 2012-11-28 Arpi Matossian-Rogers Peptides for treatment and diagnosis of autoimmunedisease
WO2007022287A2 (en) 2005-08-15 2007-02-22 Vegenics Limited Modified vegf and pdgf with improved angiogenic properties
AU2006291165B2 (en) 2005-09-12 2013-03-14 The Ohio State University Research Foundation Compositions and methods for the diagnosis and therapy of BCL2-associated cancers
US20080200408A1 (en) * 2005-09-30 2008-08-21 Mccormack Kenneth Deletion mutants of tetrodotoxin-resistant sodium channel alpha subunit
US7972813B2 (en) * 2005-09-30 2011-07-05 Vertex Pharmaceuticals Incorporated Tetrodotoxin-resistant sodium channel alpha subunit
GB2432366B (en) * 2005-11-19 2007-11-21 Alligator Bioscience Ab A method for in vitro molecular evolution of protein function
CA2524619A1 (en) * 2005-11-22 2007-05-22 Ottawa Health Research Institute Novel stem cells, nucleotide sequences and proteins therefrom
ATE480559T1 (en) 2005-12-14 2010-09-15 Licentia Ltd USES OF A NEUROTROPHIC FACTOR
WO2007089375A2 (en) 2005-12-22 2007-08-09 Exegenics, Inc. D/B/A Opko Health, Inc. Compositions and methods for regulating complement system
NZ569883A (en) 2005-12-28 2011-12-22 Translational Therapeutics Inc Use of triazole containing compounds for inhibiting 4E activity and proliferation of a cell
EP2487252B1 (en) 2006-01-05 2014-10-15 The Ohio State University Research Foundation MicroRNA-based methods for the diagnosis of colon cancer
CN103993082B (en) 2006-01-05 2017-01-11 俄亥俄州立大学研究基金会 Microrna-based methods and compositions for the diagnosis, prognosis and treatment of lung cancer
JP5490413B2 (en) 2006-01-05 2014-05-14 ジ・オハイオ・ステイト・ユニバーシティ・リサーチ・ファウンデイション Abnormal microRNA expression in pancreatic endocrine and acinar tumors
US20090214496A1 (en) * 2006-01-30 2009-08-27 Licentia Ltd. Bmx/etk tyrosine kinase gene therapy materials and methods
US8229398B2 (en) * 2006-01-30 2012-07-24 Qualcomm Incorporated GSM authentication in a CDMA network
EP2522748A1 (en) 2006-03-02 2012-11-14 The Ohio State University MicroRNA expression profile associated with pancreatic cancer
CA2647769C (en) 2006-03-20 2015-05-26 Japan Science And Technology Agency Control of intracellular target molecule by ip3 receptor-binding protein
EP2369012A1 (en) 2006-03-20 2011-09-28 The Ohio State University Research Foundation Micro-RNA fingerprints during human megakaryocytopoiesis
AU2007233109B2 (en) 2006-03-31 2010-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 gene
WO2007119180A2 (en) 2006-04-07 2007-10-25 Neuro Therapeutics Ab Survival and development of neural cells
EP2013222B1 (en) * 2006-04-28 2013-02-13 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of a gene from the jc virus
US7723564B2 (en) 2006-05-10 2010-05-25 Board Of Regents Of The University Of Nebraska Compositions and methods for modulation of KSR1 and KSR2 interactions
PL2194128T3 (en) 2006-05-11 2012-12-31 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of the PCSK9 gene
ES2497641T3 (en) 2006-05-17 2014-09-23 The Ludwig Institute For Cancer Research Direction to the regulation of VEGF-B of fatty acid transporters to modulate human diseases
BRPI0712034A2 (en) * 2006-05-19 2012-01-10 Alnylam Pharmaceuticals Inc aha rnai modulation and therapeutic uses thereof
EP2192200B1 (en) 2006-05-22 2012-10-24 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of IKK-B gene
US8598333B2 (en) * 2006-05-26 2013-12-03 Alnylam Pharmaceuticals, Inc. SiRNA silencing of genes expressed in cancer
ATE522541T1 (en) 2006-06-09 2011-09-15 Novartis Ag BACTERIAL ADHESIN CONFORMERS
EP2383341A1 (en) 2006-06-12 2011-11-02 Exegenics, Inc., D/b/a Opko Health, Inc. Compositions and methods for siRNA inhibition of angiogenesis
EP2041317A4 (en) 2006-07-13 2009-10-14 Univ Ohio State Res Found Micro-rna-based methods and compositions for the diagnosis and treatment of colon cancer-related diseases
JP2009544731A (en) 2006-07-28 2009-12-17 サノフィ−アベンティス Compositions and methods for tumor therapy
US7872118B2 (en) * 2006-09-08 2011-01-18 Opko Ophthalmics, Llc siRNA and methods of manufacture
WO2008036933A2 (en) 2006-09-21 2008-03-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the hamp gene
ES2658239T3 (en) 2006-10-19 2018-03-08 Csl Limited Interleukin-13 alpha 1 receptor high affinity antibody antagonists
PL2068922T3 (en) 2006-10-19 2012-11-30 Csl Ltd Anti-il-13r alpha 1 antibodies and their uses thereof
US20080113915A1 (en) * 2006-11-09 2008-05-15 Duke University SAPAP3 knockout mouse and clinical modeling associated with the SAPAP3 gene
AR064642A1 (en) 2006-12-22 2009-04-15 Wyeth Corp POLINUCLEOTIDE VECTOR THAT INCLUDES IT RECOMBINATING CELL THAT UNDERSTANDS THE VECTOR POLYPEPTIDE, ANTIBODY, COMPOSITION THAT UNDERSTANDS THE POLINUCLEOTIDE, VECTOR, RECOMBINATING CELL POLYPEPTIDE OR ANTIBODY, USE OF THE COMPOSITION AND A COMPOSITION AND A METHOD
WO2008097927A2 (en) * 2007-02-06 2008-08-14 Tai June Yoo Treatment and prevention of neurodegenerative diseases using gene therapy
US8841436B2 (en) * 2007-03-15 2014-09-23 University Hospitals Cleveland Medical Center Screening, diagnosing, treating and prognosis of pathophysiologic status by RNA regulation
PE20090064A1 (en) 2007-03-26 2009-03-02 Novartis Ag DOUBLE-CHAIN RIBONUCLEIC ACID TO INHIBIT THE EXPRESSION OF THE HUMAN E6AP GENE AND THE PHARMACEUTICAL COMPOSITION THAT INCLUDES IT
JP5350360B2 (en) 2007-03-29 2013-11-27 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for inhibiting the expression of genes from Ebola
EP2319926B1 (en) 2007-07-05 2016-08-31 Arrowhead Research Corporation DSRNA for treating viral infection
WO2009012263A2 (en) * 2007-07-18 2009-01-22 The Trustees Of Columbia University In The City Of New York Tissue-specific micrornas and compositions and uses thereof
ES2496172T3 (en) 2007-07-31 2014-09-18 The Ohio State University Research Foundation Methods to reverse methylation by targeted selection of DNMT3A and DNMT3B
US8465918B2 (en) 2007-08-03 2013-06-18 The Ohio State University Research Foundation Ultraconserved regions encoding ncRNAs
AU2008288806B2 (en) 2007-08-22 2014-11-27 The Ohio State University Research Foundation Methods and compositions for inducing deregulation of EphA7 and Erk phosphorylation in human acute leukemias
CN101939446B (en) 2007-09-06 2015-02-11 俄亥俄州立大学研究基金会 MicroRNA signatures in human ovarian cancer
AU2008310704B2 (en) 2007-10-11 2014-03-20 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Methods and compositions for the diagnosis and treatment of esphageal adenocarcinomas
FI20070808A0 (en) 2007-10-25 2007-10-25 Mart Saarma Split variants of GDNF and uses thereof
EP2848688A1 (en) 2007-12-10 2015-03-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor VII gene
US20090196854A1 (en) * 2008-02-04 2009-08-06 Kytos Biosystems S.A. Methods and compositions for use of crl 5803 cells for expression of biotherapeutics and encapsulated cell-based delivery
KR101397407B1 (en) 2008-03-05 2014-06-19 알닐람 파마슈티칼스 인코포레이티드 Compositions and methods for inhibiting expression of Eg5 and VEGF genes
FI20080326A0 (en) 2008-04-30 2008-04-30 Licentia Oy Neurotrophic factor MANF and its uses
US8052970B2 (en) 2008-06-30 2011-11-08 The Regents Of The University Of Michigan Lysosomal phospholipase A2 (LPLA2) activity as a diagnostic and therapeutic target for identifying and treating systemic lupus erythematosis
EP2331690B1 (en) 2008-09-02 2016-01-13 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of mutant egfr gene
EP3109321B1 (en) 2008-09-25 2019-05-01 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
EP3848461A1 (en) 2008-10-20 2021-07-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
AU2009313615B2 (en) 2008-11-05 2012-11-29 Regents Of The University Of Minnesota Multicomponent immunogenic composition for the prevention of beta-hemolytic streptococcal (BHS) disease
AU2009322279A1 (en) 2008-12-04 2011-07-14 Opko Pharmaceuticals, Llc Compositions and methods for selective inhibition of pro-angiogenic VEGF isoforms
JP5855462B2 (en) 2008-12-10 2016-02-09 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. DsRNA compositions targeting GNAQ and methods for inhibiting expression
US20120041051A1 (en) 2009-02-26 2012-02-16 Kevin Fitzgerald Compositions And Methods For Inhibiting Expression Of MIG-12 Gene
EP2406376A1 (en) 2009-03-12 2012-01-18 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
CA2755870C (en) 2009-03-20 2019-04-09 Angioblast Systems, Inc. Production of reprogrammed pluripotent cells
NO2424895T3 (en) 2009-04-27 2018-02-03
EP2424987B1 (en) * 2009-05-01 2017-11-15 CuRNA, Inc. Treatment of hemoglobin (hbf/hbg) related diseases by inhibition of natural antisense transcript to hbf/hbg
GB0908425D0 (en) 2009-05-15 2009-06-24 Medical Res Council Medical use
US9050276B2 (en) 2009-06-16 2015-06-09 The Trustees Of Columbia University In The City Of New York Autism-associated biomarkers and uses thereof
US9029338B2 (en) 2009-08-14 2015-05-12 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of a gene from the ebola virus
NZ599543A (en) 2009-10-30 2014-02-28 Ntf Therapeutics Inc Improved neurturin molecules
JP5907877B2 (en) 2009-11-02 2016-04-26 ザ、リージェンツ、オブ、ザ、ユニバーシティ、オブ、カリフォルニアThe Regents Of The University Of California Vault complex for delivery of cytokines
US8551781B2 (en) 2009-11-19 2013-10-08 The Regents Of The University Of California Vault complexes for facilitating biomolecule delivery
WO2011069155A1 (en) * 2009-12-04 2011-06-09 Opko Ophthalmics, Llc Compositions and methods for inhibition of vegf
WO2011123468A1 (en) 2010-03-29 2011-10-06 Alnylam Pharmaceuticals, Inc. Sirna therapy for transthyretin (ttr) related ocular amyloidosis
US9517250B2 (en) 2010-04-28 2016-12-13 The J. David Gladstone Institutes Methods for generating cardiomyocytes
WO2011143124A2 (en) 2010-05-10 2011-11-17 The Regents Of The University Of California Endoribonuclease compositions and methods of use thereof
CA3102008A1 (en) 2010-06-02 2011-12-08 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
EP2580328A2 (en) 2010-06-11 2013-04-17 Cellartis AB Micrornas for the detection and isolaton of human embryonic stem cell-derived cardiac cell types
JP2013539453A (en) 2010-07-09 2013-10-24 エクセリクシス, インク. Combination of kinase inhibitors for the treatment of cancer
GB201012651D0 (en) 2010-07-28 2010-09-15 Univ Edinburgh Peptides
HUE052850T2 (en) 2010-08-23 2021-05-28 Wyeth Llc Stable formulations of neisseria meningitidis rlp2086 antigens
MX362802B (en) 2010-09-10 2019-02-13 Wyeth Llc Star Non-lipidated variants of neisseria meningitidis orf2086 antigens.
CN108404115A (en) 2010-10-15 2018-08-17 纽约市哥伦比亚大学理事会 The relevant gene of obesity-and their albumen and its purposes
KR102027394B1 (en) 2010-11-02 2019-10-01 더 트러스티스 오브 콜롬비아 유니버시티 인 더 시티 오브 뉴욕 Methods for treating hair loss disorders
CN106947741A (en) 2010-11-05 2017-07-14 斯坦福大学托管董事会 Photoactivation is fitted together to opsin and its application method
JP6355335B2 (en) 2010-11-05 2018-07-11 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー Optogenetic control of reward-related behavior
EP2663868A2 (en) 2010-12-01 2013-11-20 The University of North Carolina at Chapel Hill Methods and compositions for targeting sites of neovascular growth
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
EP2649182A4 (en) 2010-12-10 2015-05-06 Alnylam Pharmaceuticals Inc Compositions and methods for increasing erythropoietin (epo) production
WO2012109495A1 (en) 2011-02-09 2012-08-16 Metabolic Solutions Development Company, Llc Cellular targets of thiazolidinediones
SG193923A1 (en) 2011-03-29 2013-11-29 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of tmprss6 gene
WO2012142526A1 (en) 2011-04-14 2012-10-18 Modiano Jaime Use of tumor fas expression to determine response to anti-cancer therapy
AU2012255143A1 (en) 2011-05-19 2014-02-20 The Regents Of The University Of Michigan Integrin alpha-2 binding agents and use thereof to inhibit cancer cell proliferation
US20140134728A1 (en) 2011-06-01 2014-05-15 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods for adjusting expression of mitochondrial genome by microrna
EP3564393A1 (en) 2011-06-21 2019-11-06 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
CA3191066A1 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
EP3388068A1 (en) 2011-06-21 2018-10-17 Alnylam Pharmaceuticals, Inc. Composition and methods for inhibition of expression of protein c (proc) genes
CN103890000B (en) 2011-06-21 2017-09-01 阿尔尼拉姆医药品有限公司 (ANGPTL3) the iRNA compositions of angiopoietin-like 3 and its application method
EP4134433A1 (en) 2011-06-23 2023-02-15 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
WO2013019857A2 (en) 2011-08-01 2013-02-07 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
EP2755681B1 (en) 2011-09-15 2018-06-27 Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center IMMUNOTHERAPY AND DIAGNOSIS OF MUCORMYCOSIS USING CotH
CN104364390B (en) 2011-10-14 2016-08-24 俄亥俄州立大学 The method relevant to ovarian cancer and material
WO2013060894A1 (en) 2011-10-27 2013-05-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment and diagnosis of atherosclerosis
US9265813B2 (en) 2011-10-27 2016-02-23 Wellstat Ophthalmics Corporation Vectors encoding rod-derived cone viability factor
WO2013079687A1 (en) 2011-11-30 2013-06-06 The Chancellor, Masters And Scholars Of The University Of Oxford Inkt cell modulators and methods of using the same
GB201120860D0 (en) 2011-12-05 2012-01-18 Cambridge Entpr Ltd Cancer immunotherapy
WO2013090356A2 (en) 2011-12-16 2013-06-20 The Board Of Trustees Of The Leland Stanford Junior University Opsin polypeptides and methods of use thereof
CA2864300A1 (en) 2012-02-16 2013-08-22 Atyr Pharma, Inc. Histidyl-trna synthetases for treating autoimmune and inflammatory diseases
ES2728077T3 (en) 2012-02-21 2019-10-22 Univ Leland Stanford Junior Compositions for the treatment of neurogenic disorders of the pelvic floor
SA115360586B1 (en) 2012-03-09 2017-04-12 فايزر انك Neisseria meningitidis compositions and methods thereof
MY198910A (en) 2012-03-09 2023-10-02 Pfizer Neisseria meningitidis compositions and methods thereof
US10039777B2 (en) 2012-03-20 2018-08-07 Neuro-Lm Sas Methods and pharmaceutical compositions of the treatment of autistic syndrome disorders
CN104334191A (en) 2012-03-29 2015-02-04 纽约市哥伦比亚大学托管会 Methods for treating hair loss disorders
US20150190532A1 (en) 2012-04-04 2015-07-09 The Trustees Of Columbia University In The City Of New York Smooth muscle specific inhibition for anti-restenotic therapy
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
WO2013153082A1 (en) 2012-04-10 2013-10-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of nonalcoholic steatohepatitis
WO2013153139A1 (en) 2012-04-11 2013-10-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment and diagnosis of acute leukemia
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
PT3241902T (en) 2012-05-25 2018-05-28 Univ California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
WO2013184209A1 (en) 2012-06-04 2013-12-12 Ludwig Institute For Cancer Research Ltd. Mif for use in methods of treating subjects with a neurodegenerative disorder
WO2014011705A1 (en) 2012-07-09 2014-01-16 The Regents Of The University Of California Vault immunotherapy
KR102165464B1 (en) 2012-07-19 2020-10-14 레드우드 바이오사이언스 인코포레이티드 Antibody specific for cd22 and methods of use thereof
US20140037599A1 (en) * 2012-08-03 2014-02-06 The Trustees Of The University Of Pennsylvania Compositions and Methods of Treating T Cell Deficiency
HUE048780T2 (en) 2012-08-16 2020-09-28 Ipierian Inc Methods of treating a tauopathy
CA2884245C (en) 2012-09-06 2023-03-14 The University Of Chicago Antisense polynucleotides to induce exon skipping and methods of treating dystrophies
CA2889170C (en) 2012-10-25 2021-09-07 True North Therapeutics, Inc. Anti-complement c1s antibodies and uses thereof
CN104884088B (en) 2012-11-02 2018-06-15 美国比奥维拉迪维股份有限公司 Anticomplement C1s antibody and its purposes
EP2922476B1 (en) 2012-11-21 2019-10-09 Circuit Therapeutics, Inc. System for optogenetic therapy
WO2014089313A1 (en) 2012-12-05 2014-06-12 Alnylam Pharmaceuticals PCSK9 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP2956557B1 (en) 2013-02-14 2018-07-11 The J. David Gladstone Institutes Compositions and methods of use thereof for identifying anti-viral agents
CA2901115A1 (en) 2013-02-15 2014-08-21 The Regents Of The University Of California Heterodimeric conditionally active chimeric antigen receptor and methods of use thereof
SG11201505940WA (en) 2013-02-18 2015-08-28 Vegenics Pty Ltd Ligand binding molecules and uses thereof
CA2903716C (en) 2013-03-08 2019-04-09 Pfizer Inc. Immunogenic fusion polypeptides
CA2940513C (en) 2013-03-11 2023-08-15 University Of Florida Research Foundation, Inc. Delivery of card protein as therapy for ocular inflammation
MX360560B (en) 2013-03-13 2018-11-07 Geneweave Biosciences Inc Non-replicative transduction particles and transduction particle-based reporter systems.
US9481903B2 (en) 2013-03-13 2016-11-01 Roche Molecular Systems, Inc. Systems and methods for detection of cells using engineered transduction particles
PE20160046A1 (en) 2013-03-14 2016-02-14 Alnylam Pharmaceuticals Inc COMPOSITION OF RNAi AGAINST THE C5 COMPONENT OF THE COMPLEMENT AND METHODS FOR ITS USE
WO2014144409A1 (en) 2013-03-15 2014-09-18 The Board Of Trustees Of The Leland Stanford Junior University Optogenetic control of behavioral state
JP2016515508A (en) 2013-03-15 2016-05-30 ザ・トラスティーズ・オブ・コロンビア・ユニバーシティ・イン・ザ・シティ・オブ・ニューヨーク Fusion protein and method thereof
EP3708184A1 (en) 2013-03-27 2020-09-16 The General Hospital Corporation Methods and agents for treating alzheimer s disease
US10220092B2 (en) 2013-04-29 2019-03-05 The Board Of Trustees Of The Leland Stanford Junior University Devices, systems and methods for optogenetic modulation of action potentials in target cells
SG10201913872XA (en) 2013-05-22 2020-03-30 Alnylam Pharmaceuticals Inc SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
SG11201510565TA (en) 2013-05-22 2016-01-28 Alnylam Pharmaceuticals Inc Tmprss6 irna compositions and methods of use thereof
WO2014191630A2 (en) 2013-05-28 2014-12-04 Helsingin Yliopisto Non-human animal model encoding a non-functional manf gene
AU2014272024B2 (en) 2013-05-29 2018-03-15 Vybion, Inc. Single chain intrabodies that alter huntingtin mutant degradation
WO2014197535A1 (en) 2013-06-04 2014-12-11 Virginia Commonwealth University Recombinant cancer therapeutic cytokine
WO2014197599A1 (en) 2013-06-04 2014-12-11 The Johns Hopkins University Peg-prom mediated surface expression of avidin/streptavidin
WO2014197586A1 (en) 2013-06-04 2014-12-11 Virginia Commonwealth University Mda-9/syntenin promoter to image and treat metastatic cancer cells
EP3003393A4 (en) 2013-06-04 2017-01-11 The Johns Hopkins University Tripartite cancer theranostic nucleic acid constructs
EP3003022B1 (en) 2013-06-04 2017-11-22 Virginia Commonwealth University Use of a truncated ccn1 promoter for cancer diagnostics, therapeutics and theranostics
PL3007726T3 (en) 2013-06-10 2021-01-11 Ipierian, Inc. Methods of treating a tauopathy
EP3013424A4 (en) 2013-06-25 2017-03-29 University of Canberra Methods and compositions for modulating cancer stem cells
US10307609B2 (en) 2013-08-14 2019-06-04 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods for controlling pain
RU2662968C2 (en) 2013-09-08 2018-07-31 Пфайзер Инк. Immunogenic composition for neisseria meningitidis (options)
ES2851724T3 (en) 2013-09-18 2021-09-08 Epiaxis Therapeutics Pty Ltd Stem cell modulation
EP3052626A1 (en) 2013-10-02 2016-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
TW202310853A (en) 2013-10-04 2023-03-16 美國西奈山伊坎醫學院 Compositions and methods for inhibiting expression of the alas1 gene
JP2016537028A (en) 2013-11-18 2016-12-01 クリスパー セラピューティクス アーゲー CRISPR-CAS System Materials and Methods
WO2015089277A1 (en) 2013-12-12 2015-06-18 The Regents Of The University Of California Methods and compositions for modifying a single stranded target nucleic acid
CA2931090A1 (en) 2013-12-12 2015-06-18 Alnylam Pharmaceuticals, Inc. Complement component irna compositions and methods of use thereof
EP3079727A1 (en) 2013-12-12 2016-10-19 INSERM - Institut National de la Santé et de la Recherche Médicale Methods for the prevention and treatment of diabetic cardiomyopathy using mir-424/322
US9682123B2 (en) 2013-12-20 2017-06-20 The Trustees Of Columbia University In The City Of New York Methods of treating metabolic disease
WO2015123264A1 (en) 2014-02-11 2015-08-20 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
WO2015127094A1 (en) 2014-02-19 2015-08-27 University Of Florida Research Foundation, Inc. Delivery of nrf2 as therapy for protection against reactive oxygen species
EP3581580A1 (en) 2014-03-28 2019-12-18 The Board of Trustees of the Leland Stanford Junior University Engineered light-activated anion channel proteins and methods of use thereof
TW201607559A (en) 2014-05-12 2016-03-01 阿尼拉製藥公司 Methods and compositions for treating a SERPINC1-associated disorder
EA201692370A1 (en) 2014-05-22 2017-03-31 Элнилэм Фармасьютикалз, Инк. COMPOSITIONS of mRNA ANGIOTENZINOGENA (AGT) AND METHODS OF THEIR USE
EP3151866B1 (en) 2014-06-09 2023-03-08 Voyager Therapeutics, Inc. Chimeric capsids
PT3157552T (en) 2014-06-18 2020-01-22 Albert Einstein College Medicine Inc Syntac polypeptides and uses thereof
PL3177640T3 (en) 2014-08-08 2020-11-02 The Board Of Trustees Of The Leland Stanford Junior University High affinity pd-1 agents and methods of use
WO2016029262A1 (en) 2014-08-25 2016-03-03 University Of Canberra Compositions for modulating cancer stem cells and uses therefor
CN107002046B (en) 2014-08-25 2021-05-11 经纬生物科技有限公司 Non-replicating transduction particles and transduction particle-based reporter systems
MX2017002935A (en) 2014-09-07 2017-05-30 Selecta Biosciences Inc Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses.
CN107295802B (en) 2014-09-24 2021-06-29 希望之城 Adeno-associated virus vector variants for efficient genome editing and methods thereof
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
CA2966620A1 (en) 2014-11-05 2016-05-12 Voyager Therapeutics, Inc. Aadc polynucleotides for the treatment of parkinson's disease
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
RU2719192C2 (en) 2014-11-14 2020-04-17 Вояджер Терапьютикс, Инк. Modulating polynucleotides
CN114717264A (en) 2014-11-14 2022-07-08 沃雅戈治疗公司 Compositions and methods for treating Amyotrophic Lateral Sclerosis (ALS)
CN113846101A (en) 2014-11-17 2021-12-28 阿尔尼拉姆医药品有限公司 Apolipoprotein C3(APOC3) iRNA compositions and methods of use thereof
US11697825B2 (en) 2014-12-12 2023-07-11 Voyager Therapeutics, Inc. Compositions and methods for the production of scAAV
EP3778644A3 (en) 2014-12-23 2021-06-02 The Trustees of Columbia University in the City of New York Fgfr-tacc fusion proteins and methods thereof
CA2976445A1 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
US10888611B2 (en) 2015-02-19 2021-01-12 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
KR102004076B1 (en) 2015-03-13 2019-07-25 더 잭슨 래보라토리 Three-component CRISPR / CAS complex system and its uses
SG10201909180SA (en) 2015-04-06 2019-11-28 Bioverativ Usa Inc Humanized anti-c1s antibodies and methods of use thereof
WO2016164371A1 (en) 2015-04-07 2016-10-13 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Methods for inducing cell division of postmitotic cells
EP3283500B1 (en) 2015-04-08 2020-11-11 The University of Chicago Compositions and methods for correcting limb girdle muscular dystrophy type 2c using exon skipping
US10729790B2 (en) 2015-05-26 2020-08-04 Salk Institute For Biological Studies Motor neuron-specific expression vectors
EP3303634B1 (en) 2015-06-03 2023-08-30 The Regents of The University of California Cas9 variants and methods of use thereof
EP3307316A1 (en) 2015-06-12 2018-04-18 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2017023861A1 (en) 2015-08-03 2017-02-09 The Regents Of The University Of California Compositions and methods for modulating abhd2 activity
EP3344654B1 (en) 2015-09-02 2020-10-21 Immutep S.A.S. Anti-lag-3 antibodies
WO2017044807A2 (en) 2015-09-09 2017-03-16 The Trustees Of Columbia University In The City Of New York Reduction of er-mam-localized app-c99 and methods of treating alzheimer's disease
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
US10188750B1 (en) 2015-10-23 2019-01-29 University Of South Florida Self-replicating cell selective gene delivery compositions, methods, and uses thereof
US10983110B2 (en) 2015-12-02 2021-04-20 Voyager Therapeutics, Inc. Assays for the detection of AAV neutralizing antibodies
MX2018008416A (en) 2016-01-08 2019-11-11 Univ California Conditionally active heterodimeric polypeptides and methods of use thereof.
US10478503B2 (en) 2016-01-31 2019-11-19 University Of Massachusetts Branched oligonucleotides
JP2019509997A (en) 2016-02-17 2019-04-11 ザ チルドレンズ メディカル センター コーポレーション FFA1 (GPR40) as a therapeutic target for neuroangiogenic diseases or disorders
AU2017225787B2 (en) 2016-03-03 2021-09-23 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11325948B2 (en) 2016-03-19 2022-05-10 Exuma Biotech Corp. Methods and compositions for genetically modifying lymphocytes to express polypeptides comprising the intracellular domain of MPL
US11111505B2 (en) 2016-03-19 2021-09-07 Exuma Biotech, Corp. Methods and compositions for transducing lymphocytes and regulating the activity thereof
SG11201807286WA (en) 2016-03-19 2018-10-30 F1 Oncology Inc Methods and compositions for transducing lymphocytes and regulated expansion thereof
WO2020047527A2 (en) 2018-09-02 2020-03-05 F1 Bioventures, Llc Methods and compositions for genetically modifying lymphocytes in blood or in enriched pbmcs
IL262010B (en) 2016-04-04 2022-09-01 Bioverativ Usa Inc Anti-complement factor bb antibodies and uses thereof
WO2017182981A1 (en) 2016-04-20 2017-10-26 Washington University Ppar agonist or lxr agonist for use in the treatment of systemic lupus erythematosus by modulation of lap activity
US11326182B2 (en) 2016-04-29 2022-05-10 Voyager Therapeutics, Inc. Compositions for the treatment of disease
WO2017189964A2 (en) 2016-04-29 2017-11-02 Voyager Therapeutics, Inc. Compositions for the treatment of disease
CN109689096A (en) 2016-05-18 2019-04-26 阿尔伯特爱因斯坦医学院公司 Variant PD-L1 polypeptide, T cell modulability multimeric polypeptide and its application method
JP7066635B2 (en) 2016-05-18 2022-05-13 ボイジャー セラピューティクス インコーポレイテッド Modulatory polynucleotide
BR112018073606A2 (en) 2016-05-18 2019-02-26 Cue Biopharma, Inc. T-cell modulating multimeric polypeptides and methods of using them
US20190256845A1 (en) 2016-06-10 2019-08-22 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT C5 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
AU2017292936B2 (en) 2016-07-08 2024-02-01 Exuma Biotech, Corp. Methods and compositions for transducing lymphocytes and regulating the activity thereof
JP2019531787A (en) 2016-08-30 2019-11-07 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Biomedical targeting and delivery method and apparatus and system for performing the same
US20190218261A1 (en) 2016-09-13 2019-07-18 The Jackson Laboratory Targeted enhanced dna demethylation
WO2018057648A1 (en) 2016-09-20 2018-03-29 The Board Of Trustees Of The Leland Stanford Junior University Peptide regulators of mitochondrial fusion and methods of use
EP3299460A1 (en) 2016-09-26 2018-03-28 Johann Wolfgang Goethe-Universität Frankfurt am Main Novel compounds and methods for modulating ubiquitination
CN110300520B (en) 2016-10-12 2022-10-04 美国比奥维拉迪维股份有限公司 anti-C1 s antibodies and methods of use thereof
US11154591B2 (en) 2016-10-14 2021-10-26 The Trustees Of Columbia University In The City Of New York Methods of treating alcohol abuse disorder
US11332713B2 (en) 2016-11-16 2022-05-17 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
TWI790217B (en) 2016-12-16 2023-01-21 美商阿尼拉製藥公司 METHODS FOR TREATING OR PREVENTING TTR-ASSOCIATED DISEASES USING TRANSTHYRETIN (TTR) iRNA COMPOSITIONS
PT3558339T (en) 2016-12-22 2024-03-15 Cue Biopharma Inc T-cell modulatory multimeric polypeptides and methods of use thereof
WO2018129474A1 (en) 2017-01-09 2018-07-12 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
WO2018136566A1 (en) 2017-01-18 2018-07-26 F1 Oncology, Inc. Methods of transducing and expanding immune cells and uses thereof
MX2019008503A (en) 2017-01-18 2019-09-13 F1 Oncology Inc Chimeric antigen receptors against axl or ror2 and methods of use thereof.
KR102567845B1 (en) 2017-01-31 2023-08-17 화이자 인코포레이티드 Neisseria meningitidis compositions and methods thereof
AU2018226884A1 (en) 2017-03-03 2019-10-10 Exuma Biotech, Corp. Methods and compositions for transducing and expanding lymphocytes and regulating the activity thereof
WO2018204803A1 (en) 2017-05-05 2018-11-08 Voyager Therapeutics, Inc. Compositions and methods of treating huntington's disease
WO2018204786A1 (en) 2017-05-05 2018-11-08 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (als)
JOP20190269A1 (en) 2017-06-15 2019-11-20 Voyager Therapeutics Inc Aadc polynucleotides for the treatment of parkinson's disease
GB201710973D0 (en) 2017-07-07 2017-08-23 Avacta Life Sciences Ltd Scaffold proteins
AU2018302016A1 (en) 2017-07-17 2020-02-06 The Regents Of The University Of California Trajectory array guide system
TWI722310B (en) 2017-08-03 2021-03-21 美商航海家醫療公司 Compositions and methods for delivery of aav
EP3679064A4 (en) 2017-09-07 2021-06-02 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptide with conjugation sites and methods of use thereof
IL273119B2 (en) 2017-09-08 2023-10-01 Maverick Therapeutics Inc Constrained conditionally activated binding proteins
CN111315773A (en) 2017-09-08 2020-06-19 马弗里克治疗公司 Conditionally active binding moieties comprising an Fc region
KR20230110812A (en) 2017-10-10 2023-07-25 난트바이오 인코포레이티드 Modified ec7 cells having low toxicity to viral production payloads
WO2019075220A1 (en) 2017-10-11 2019-04-18 Bioverativ Usa Inc. Methods of inducing complement activity
CN111479924A (en) 2017-10-16 2020-07-31 沃雅戈治疗公司 Treatment of amyotrophic lateral sclerosis (A L S)
WO2019079242A1 (en) 2017-10-16 2019-04-25 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
SG11202002737PA (en) * 2017-10-18 2020-05-28 Hope City Adeno-associated virus compositions for restoring hbb gene function and methods of use thereof
AU2018360697A1 (en) 2017-11-01 2020-05-14 Alnylam Pharmaceuticals, Inc. Complement component C3 iRNA compositions and methods of use thereof
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
JP2021506251A (en) 2017-12-14 2021-02-22 クリスパー セラピューティクス アーゲー New RNA programmable endonuclease system, as well as its use in genome editing and other applications
US11008602B2 (en) 2017-12-20 2021-05-18 Roche Molecular Systems, Inc. Non-replicative transduction particles and transduction particle-based reporter systems
WO2019139896A1 (en) 2018-01-09 2019-07-18 Cue Biopharma, Inc. Multimeric t-cell modulatory polypeptides and methods of use thereof
US10610606B2 (en) 2018-02-01 2020-04-07 Homology Medicines, Inc. Adeno-associated virus compositions for PAH gene transfer and methods of use thereof
US11667949B2 (en) 2018-02-15 2023-06-06 The Trustees Of Princeton University Reporter construct and biosensor for interferon second messenger 2-5A
JP7244547B2 (en) 2018-02-19 2023-03-22 ホモロジー・メディシンズ・インコーポレイテッド Adeno-associated virus compositions and methods of use thereof for restoring F8 gene function
CA3093915A1 (en) 2018-03-15 2019-09-19 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
EP3768834A1 (en) 2018-03-19 2021-01-27 CRISPR Therapeutics AG Novel rna-programmable endonuclease systems and uses thereof
JP2021533744A (en) 2018-08-09 2021-12-09 マベリック セラピューティクス, インコーポレイテッドMaverick Therapeutics, Inc. Methods for co-expressing and purifying conditionally activated binding proteins
EP3837367A1 (en) 2018-08-16 2021-06-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
CN110857440B (en) 2018-08-23 2021-02-19 武汉纽福斯生物科技有限公司 Recombinant human II type mitochondrial dynamic protein sample GTP enzyme gene sequence and application thereof
EP3843774A4 (en) 2018-08-30 2022-08-31 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with myocardin and ascl1
CN111118016B (en) 2018-10-30 2022-04-15 上海朗昇生物科技有限公司 Gene therapy vector for treating retinitis pigmentosa diseases
WO2020104649A2 (en) 2018-11-23 2020-05-28 Sanofi Novel rna compositions and methods for inhibiting angptl8
TW202039542A (en) 2018-12-19 2020-11-01 美商庫爾生物製藥有限公司 Multimeric t-cell modulatory polypeptides and methods of use thereof
JP2022515451A (en) 2018-12-27 2022-02-18 エフ.ホフマン-ラ ロシュ アーゲー Reporter system based on non-replicating and transduced particles for the detection of Acinetobacter Baumanni
US11572595B2 (en) 2018-12-31 2023-02-07 Roche Molecular Systems, Inc. Non-replicative transduction particles with one or more non-native tail fibers and transduction particle-based reporter systems
EP3934762A1 (en) 2019-03-05 2022-01-12 Takeda Pharmaceutical Company Limited Constrained conditionally activated binding proteins
CN114173876A (en) 2019-03-05 2022-03-11 武田药品工业有限公司 Conditionally active binding proteins containing Fc regions and moieties targeting tumor antigens
EP3935080A4 (en) 2019-03-06 2023-04-05 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
GB2595606B (en) 2019-03-07 2022-09-21 Univ California CRISPR-Cas effector polypeptides and methods of use thereof
SG11202109741VA (en) 2019-03-12 2021-10-28 Crispr Therapeutics Ag Novel high fidelity rna-programmable endonuclease systems and uses thereof
CN111718418B (en) 2019-03-19 2021-08-27 华东师范大学 Fusion protein for enhancing gene editing and application thereof
KR20220004653A (en) 2019-04-02 2022-01-11 켄조케티 바이오테크놀러지 인코포레이티드 Effluent pump-cancer antigen multispecific antibodies and compositions, reagents, kits and methods related thereto
EP3956474A1 (en) 2019-04-18 2022-02-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment and prognosis of cancer
EP3736330A1 (en) 2019-05-08 2020-11-11 European Molecular Biology Laboratory Modified adeno-associated virus (aav) particles for gene therapy
CN112180094A (en) 2019-07-04 2021-01-05 上海东慈生物科技有限公司 Application of DKK1 inhibitor in preventing and/or treating tumor cachexia and diabetes-associated diseases
US20230137971A1 (en) 2019-07-11 2023-05-04 Tenaya Therapeutics Inc. Cardiac cell reprogramming with micrornas and other factors
EP4003508A1 (en) 2019-07-31 2022-06-01 Memorial Sloan Kettering Cancer Center Perfusion modulated tumor dose sculpting with single dose radiotherapy
AU2020328507A1 (en) 2019-08-12 2022-03-17 Purinomia Biotech, Inc. Methods and compositions for promoting and potentiating T-cell mediated immune responses through ADCC targeting of CD39 expressing cells
EP4039801A4 (en) 2019-08-16 2023-10-18 Center For Excellence In Brain Science And Intelligence Technology, Chinese Academy Of Sciences Application of ptbp1 inhibitor in preventing and/or treating nervous system disease related to functional neuronal death
CN115176011A (en) 2019-08-27 2022-10-11 赛诺菲 Compositions and methods for inhibiting PCSK9
JP2022546570A (en) 2019-09-03 2022-11-04 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for inhibiting expression of the LECT2 gene
EP4038189A1 (en) 2019-10-04 2022-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
TW202128775A (en) 2019-10-16 2021-08-01 英商阿法克塔生命科學有限公司 Pd-l1 inhibitor - tgfβ inhibitor bispecific drug moieties
US20230040920A1 (en) 2019-11-01 2023-02-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
TW202132568A (en) 2019-11-13 2021-09-01 美商阿尼拉製藥公司 Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
WO2021113736A1 (en) 2019-12-05 2021-06-10 Massachusetts Institute Of Technology Single-domain antibody to chloramphenicol
EP4085156B1 (en) 2019-12-31 2023-10-25 F. Hoffmann-La Roche AG Quantitative pcr screening of inducible prophage from bacterial isolates
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
WO2021163066A1 (en) 2020-02-10 2021-08-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
AU2021230476A1 (en) 2020-03-02 2022-10-20 Tenaya Therapeutics, Inc. Gene vector control by cardiomyocyte-expressed microRNAs
GB202004498D0 (en) 2020-03-27 2020-05-13 Ucl Business Ltd Activity-dependent gene therapy for neurological disorders
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
MX2022012209A (en) 2020-03-30 2022-10-27 Wuhan Neurophth Biotechnology Ltd Company Expression vector of human nuclear factor e2-related factor 2 and application of expression vector.
BR112022020145A2 (en) 2020-04-06 2023-01-03 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR SILENCING THE MYOC EXPRESSION
TW202204617A (en) 2020-04-07 2022-02-01 美商艾爾妮蘭製藥公司 Compositions and methods for silencing scn9a expression
WO2021237097A1 (en) 2020-05-21 2021-11-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
CN116529267A (en) 2020-06-04 2023-08-01 肯乔克蒂生物技术股份有限公司 ABCG2 efflux pump-cancer antigen multispecific antibodies and related compositions, reagents, kits and methods
GB202101299D0 (en) 2020-06-09 2021-03-17 Avacta Life Sciences Ltd Diagnostic polypetides and methods
GB202010981D0 (en) 2020-07-16 2020-09-02 Ucl Business Ltd Gene therapy for neuromuscular and neuromotor disorders
WO2022032085A1 (en) 2020-08-07 2022-02-10 The Jackson Laboratory Targeted sequence insertion compositions and methods
US11781156B2 (en) 2020-10-09 2023-10-10 Tenaya Therapeutics, Inc. Plakophillin-2 gene therapy methods and compositions
WO2022079221A1 (en) 2020-10-16 2022-04-21 Sanofi Rna compositions and methods for inhibiting lipoprotein(a)
US20230383294A1 (en) 2020-10-16 2023-11-30 Sanofi Novel rna compositions and methods for inhibiting angptl3
AU2021365822A1 (en) 2020-10-21 2023-06-08 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
JP2023548432A (en) 2020-11-11 2023-11-16 ヨーロピアン モレキュラー バイオロジー ラボラトリー Modified virus particles for gene therapy
TW202237150A (en) 2020-12-01 2022-10-01 美商艾拉倫製藥股份有限公司 Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
WO2022187289A1 (en) 2021-03-01 2022-09-09 Exuma Biotech Corp. Methods and compositions for the delivery of retroviral particles
WO2022234003A1 (en) 2021-05-07 2022-11-10 Avacta Life Sciences Limited Cd33 binding polypeptides with stefin a protein
BR112023023768A2 (en) 2021-06-11 2024-02-27 Bayer Ag TYPE V RNA PROGRAMMABLE ENDONUCLEASE SYSTEMS
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
KR20240026203A (en) 2021-06-30 2024-02-27 알닐람 파마슈티칼스 인코포레이티드 Methods and compositions for treating angiotensinogen (AGT)-related disorders
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
TW202332694A (en) 2021-10-07 2023-08-16 英商阿凡克塔生命科學公司 Serum half-life extended pd-l1 binding polypeptides
TW202334196A (en) 2021-10-07 2023-09-01 英商阿法克塔生命科學有限公司 Pd-l1 binding polypeptides
WO2023102388A1 (en) 2021-11-30 2023-06-08 Sanofi Pasteur Inc. Human metapneumovirus viral vector-based vaccines
WO2023114658A1 (en) 2021-12-13 2023-06-22 Kenjockety Biotechnology, Inc. Anti-abcb1 antibodies
WO2023118068A1 (en) 2021-12-23 2023-06-29 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2023131682A1 (en) 2022-01-06 2023-07-13 Ucl Business Ltd Endogenous gene regulation to treat neurological disorders and diseases
GB202201744D0 (en) 2022-02-10 2022-03-30 Ucl Business Ltd Treatment of acquired focal epilepsy
WO2023159220A1 (en) 2022-02-18 2023-08-24 Kenjockety Biotechnology, Inc. Anti-cd47 antibodies
WO2023168305A1 (en) 2022-03-01 2023-09-07 Exuma Biotech Corp. Viral particles with membrane-bound hyaluronidase
WO2023222829A1 (en) 2022-05-17 2023-11-23 Centre Hospitalier Universitaire Vaudois Designed biosensors for enhanced t cell therapy
WO2023237587A1 (en) 2022-06-10 2023-12-14 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
EP4299739A1 (en) 2022-06-30 2024-01-03 Inari Agriculture Technology, Inc. Compositions, systems, and methods for genome editing
WO2024005864A1 (en) 2022-06-30 2024-01-04 Inari Agriculture Technology, Inc. Compositions, systems, and methods for genome editing
EP4299733A1 (en) 2022-06-30 2024-01-03 Inari Agriculture Technology, Inc. Compositions, systems, and methods for genome editing
WO2024005863A1 (en) 2022-06-30 2024-01-04 Inari Agriculture Technology, Inc. Compositions, systems, and methods for genome editing
WO2024044659A1 (en) 2022-08-24 2024-02-29 Tectonic Therapeutic, Inc. Constitutively active g protein-coupled receptor compositions and methods of use thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5139941A (en) * 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
WO1988008450A1 (en) * 1987-05-01 1988-11-03 Birdwell Finlayson Gene therapy for metabolite disorders
AU7906691A (en) * 1990-05-23 1991-12-10 United States of America, as represented by the Secretary, U.S. Department of Commerce, The Adeno-associated virus (aav)-based eucaryotic vectors
US5173414A (en) * 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors

Also Published As

Publication number Publication date
JPH06504680A (en) 1994-06-02
KR930703453A (en) 1993-11-30
EP0566732B1 (en) 2003-04-02
US5252479A (en) 1993-10-12
PT101040A (en) 1994-05-31
DE69232983T2 (en) 2003-11-13
IL103677A0 (en) 1993-04-04
PT101040B (en) 2003-06-30
DK0566732T3 (en) 2003-07-21
ATE236263T1 (en) 2003-04-15
DE69232983D1 (en) 2003-05-08
ES2192555T3 (en) 2003-10-16
WO1993009239A1 (en) 1993-05-13
AU657829B2 (en) 1995-03-23
CA2098483A1 (en) 1993-05-09
EP0566732A1 (en) 1993-10-27
JP2003174896A (en) 2003-06-24
AU3134093A (en) 1993-06-07

Similar Documents

Publication Publication Date Title
CA2098483C (en) Safe adeno-associated virus 2 vector for gene therapy
US6261834B1 (en) Vector for gene therapy
AU688428B2 (en) Generation of high titers of recombinant AAV vectors
US6951753B2 (en) Helper-free stocks of recombinant adeno-associated virus vectors
Fisher et al. Recombinant adeno-associated virus for muscle directed gene therapy
Ponnazhagan et al. Adeno-associated virus 2-mediated gene transfer in vivo: organ-tropism and expression of transduced sequences in mice
JP2001506133A (en) AAV split-packaging genes and cell lines containing such genes for use in producing recombinant AAV vectors
EP0644944A1 (en) Adeno-associated virus with inverted terminal repeat sequences as promoter
WO1998027204A9 (en) Aav split-packaging genes and cell lines comprising such genes for use in the production of recombinant aav vectors
Walsh et al. Gene therapy for human hemoglobinopathies
CA2269661A1 (en) Recombinase-activatable aav packaging cassettes for use in the production of aav vectors
CA2274146C (en) Genetic modification of primate hemopoietic repopulating stem cells
EP1027451A1 (en) Chimeric parvovirus-based recombinant vector system that specifically targets the erythroid lineage
AU693317B2 (en) Retroviral vectors for expression in embryonic cells
WO2023220035A1 (en) Erythroparvovirus compositions and methods for gene therapy
WO2023220040A1 (en) Erythroparvovirus with a modified capsid for gene therapy
Washington et al. Gene Therapy for Nonneoplastic Hematologic and Histiocytic Disorders
Kroner‐Lux et al. Delivery Systems for Gene Therapy: Adeno‐Associated Virus
AU7211201A (en) Genetic modification of primate hemopoietic repopulating stem cells

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed
MKLA Lapsed

Effective date: 20061106