US20160272976A1 - Products and methods for treatment of familial amyotrophic lateral sclerosis - Google Patents

Products and methods for treatment of familial amyotrophic lateral sclerosis Download PDF

Info

Publication number
US20160272976A1
US20160272976A1 US14/914,861 US201414914861A US2016272976A1 US 20160272976 A1 US20160272976 A1 US 20160272976A1 US 201414914861 A US201414914861 A US 201414914861A US 2016272976 A1 US2016272976 A1 US 2016272976A1
Authority
US
United States
Prior art keywords
sod1
seq
shrna
aav
aav9
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.)
Abandoned
Application number
US14/914,861
Other languages
English (en)
Inventor
Brian K. Kaspar
Kevin Foust
Don W. Cleveland
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.)
Ludwig Institute for Cancer Research Ltd
Research Institute at Nationwide Childrens Hospital
Original Assignee
Ludwig Institute for Cancer Research Ltd
Research Institute at Nationwide Childrens Hospital
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 Ludwig Institute for Cancer Research Ltd, Research Institute at Nationwide Childrens Hospital filed Critical Ludwig Institute for Cancer Research Ltd
Priority to US14/914,861 priority Critical patent/US20160272976A1/en
Assigned to LUDWIG INSTITUTE FOR CANCER RESEARCH, LTD reassignment LUDWIG INSTITUTE FOR CANCER RESEARCH, LTD ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CLEVELAND, DON W.
Assigned to RESEARCH INSTITUTE AT NATIONWIDE CHILDREN'S HOSPITAL reassignment RESEARCH INSTITUTE AT NATIONWIDE CHILDREN'S HOSPITAL ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FOUST, KEVIN, KASPAR, BRIAN K.
Publication of US20160272976A1 publication Critical patent/US20160272976A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR reassignment NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: CHILDREN'S HOSPITAL (COLUMBUS)
Abandoned legal-status Critical Current

Links

Images

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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0089Oxidoreductases (1.) acting on superoxide as acceptor (1.15)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y115/00Oxidoreductases acting on superoxide as acceptor (1.15)
    • C12Y115/01Oxidoreductases acting on superoxide as acceptor (1.15) with NAD or NADP as acceptor (1.15.1)
    • C12Y115/01001Superoxide dismutase (1.15.1.1)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • C12N2310/531Stem-loop; Hairpin
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific
    • 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
    • C12N2750/14041Use of virus, viral particle or viral elements as a vector
    • C12N2750/14043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use 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

Definitions

  • the present invention relates to RNA-based methods for inhibiting the expression of the superoxide dismutase 1 (SOD-1) gene.
  • Recombinant adeno-associated viruses of the invention deliver DNAs encoding RNAs that knock down the expression of SOD-1.
  • the methods have application in the treatment of amyotrophic lateral sclerosis (ALS).
  • ALS amyotrophic lateral sclerosis
  • ALS is an adult-onset, rapidly progressive and fatal neurodegenerative disease, characterized by selective degeneration of both upper and lower motor neurons.
  • ALS is responsible for one in every 2000 deaths, affecting nearly 5 out of 100,000 individuals.
  • ALS occurs when specific nerve cells in the brain and spinal cord that control voluntary movement degenerate.
  • the loss of these motor neurons leads to progressive atrophy of skeletal muscles, which results in loss of muscular function resulting in paralysis, speech deficits, and death due to respiratory failure.
  • SOD1 superoxide dismutase 1
  • mutant SOD1 protein expression within motor neurons themselves contributes to disease onset and early disease progression 6 , as does mutant synthesis in NG2 + cells' that are precursors to oligodendrocytes.
  • mutant SOD1 protein expression in microglia and astrocytes significantly drives rapid disease progression 6,8 , findings which have lead to the conclusion that ALS pathophysiology is non-cell autonomous 3 .
  • astrocytes have been found to be toxic to motor neurons in multiple in vitro models where mutant forms of human SOD1 were overexpressed 9-11 .
  • a recent study derived astrocytes from post-mortem spinal cords of ALS patients with or without SOD1 mutations. In all cases, astrocytes from sporadic ALS patients were as toxic to motor neurons as astrocytes carrying genetic mutations in SOD1 12 . Even more strikingly, reduction of SOD1 in astrocytes derived from both sporadic and familial ALS patients decreased astrocyte-derived toxicity that is selective for motor, but not GABA, neurons.
  • riluzole is the only drug currently approved by the FDA as a therapy for ALS, providing a modest survival benefit 21 .
  • attempts at improving therapy by reducing synthesis of SOD1 have been the focus of multiple therapeutic development approaches.
  • Antisense oligonucleotides and viral delivered RNA interference (RNAi) were tested in rat 22 and mouse models 23-25 that develop fatal paralysis from overexpressing human SOD1 G93A .
  • Antisense oligonucleotides infused at disease onset produced SOD1 reduction and a modest slowing of disease progression 22 .
  • Adeno-associated virus (AAV) vectors have been used in a number of recent clinical trials for treatment of neurological disorders [Kaplitt et al., Lancet 369: 2097-2105 (2007); Marks et al., Lancet Neurol 7: 400-408 (2008); Worgall et al., Hum Gene Ther (2008)].
  • AAV is a replication-deficient parvovirus, the single-stranded DNA genome of which is about 4.7 kb in length including 145 nucleotide inverted terminal repeat (ITRs).
  • ITRs nucleotide inverted terminal repeat
  • AAV2 AAV serotype 2
  • Cis-acting sequences directing viral DNA replication (rep), encapsidation/packaging and host cell chromosome integration are contained within the ITRs.
  • AAV promoters Three AAV promoters (named p5, p19, and p40 for their relative map locations) drive the expression of the two AAV internal open reading frames encoding rep and cap genes.
  • the two rep promoters (p5 and p19), coupled with the differential splicing of the single AAV intron (at nucleotides 2107 and 2227), result in the production of four rep proteins (rep 78, rep 68, rep 52, and rep 40) from the rep gene.
  • Rep proteins possess multiple enzymatic properties that are ultimately responsible for replicating the viral genome.
  • the cap gene is expressed from the p40 promoter and it encodes the three capsid proteins VP1, VP2, and VP3.
  • AAV possesses unique features that make it attractive as a vector for delivering foreign DNA to cells, for example, in gene therapy.
  • AAV infection of cells in culture is noncytopathic, and natural infection of humans and other animals is silent and asymptomatic.
  • AAV infects many mammalian cells allowing the possibility of targeting many different tissues in vivo.
  • AAV transduces slowly dividing and non-dividing cells, and can persist essentially for the lifetime of those cells as a transcriptionally active nuclear episome (extrachromosomal element).
  • the AAV proviral genome is infectious as cloned DNA in plasmids which makes construction of recombinant genomes feasible.
  • the signals directing AAV replication, genome encapsidation and integration are contained within the ITRs of the AAV genome, some or all of the internal approximately 4.3 kb of the genome (encoding replication and structural capsid proteins, rep-cap) may be replaced with foreign DNA such as a gene cassette containing a promoter, a DNA of interest and a polyadenylation signal.
  • the rep and cap proteins may be provided in trans.
  • Another significant feature of AAV is that it is an extremely stable and hearty virus. It easily withstands the conditions used to inactivate adenovirus (56° to 65° C. for several hours), making cold preservation of AAV less critical. AAV may even be lyophilized. Finally, AAV-infected cells are not resistant to superinfection.
  • serotypes of AAV exist and offer varied tissue tropism.
  • Known serotypes include, for example, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11 and AAVrh74.
  • Advances in the delivery of AAV6 and AAV8 have made possible the transduction by these serotypes of skeletal and cardiac muscle following simple systemic intravenous or intraperitoneal injections. See Pacak et al., Circ. Res., 99(4): 3-9 (1006) and Wang et al., Nature Biotech., 23(3): 321-8 (2005).
  • the use of AAV to target cell types within the central nervous system has involved surgical intraparenchymal injection.
  • the present invention provides products and methods useful for reducing mutant SOD1 protein levels in subjects in need thereof.
  • the invention provides AAV-mediated delivery of RNAs including, but not limited to short hairpin RNAs, to reduce synthesis of ALS-causing human SOD1 mutants in subjects in need thereof.
  • Recombinant AAV (rAAV) contemplated by the invention include, but are not limited to, rAAV9, rAAV2 and rAAVrh74.
  • Delivery routes contemplated by the invention include, but are not limited to, systemic delivery and intrathecal delivery. Use of the methods and products of the invention is indicated, for example, in treating ALS.
  • the invention provides rAAV genomes comprising one or more AAV ITRs flanking a polynucleotide encoding one or more RNAs (including, but not limited to, small hairpin RNAs, antisense RNAs and/or microRNAs) that target mutant SOD1 polynucleotides.
  • RNAs including, but not limited to, small hairpin RNAs, antisense RNAs and/or microRNAs
  • shRNAs small hairpin RNAs
  • the shRNA-encoding polynucleotide is operatively linked to transcriptional control DNA, specifically promoter DNA that is functional in target cells.
  • Commercial providers such as Ambion Inc. (Austin, Tex.), Darmacon Inc.
  • the rAAV genome comprises a DNA encoding a SOD1 shRNA such as:
  • the rAAV genomes of the invention lack AAV rep and cap DNA.
  • AAV DNA in the rAAV genomes may be from any AAV serotype for which a recombinant virus can be derived including, but not limited to, AAV serotypes AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV-10 and AAV-11.
  • the nucleotide sequences of the genomes of the AAV serotypes are known in the art. For example, the complete genome of AAV-1 is provided in GenBank Accession No. NC_002077; the complete genome of AAV-2 is provided in GenBank Accession No.
  • AAV-9 genome is provided in Gao et al., J.
  • the AAVrh74 genome is provided in International Publication No. WO 2013/078316.
  • the invention provides DNA plasmids comprising rAAV genomes of the invention.
  • the DNA plasmids are transferred to cells permissible for infection with a helper virus of AAV (e.g., adenovirus, E1-deleted adenovirus or herpesvirus) for assembly of the rAAV genome into infectious viral particles.
  • helper virus of AAV e.g., adenovirus, E1-deleted adenovirus or herpesvirus
  • rAAV genome a rAAV genome
  • AAV rep and cap genes separate from (i.e., not in) the rAAV genome
  • helper virus functions The AAV rep and cap genes may be from any AAV serotype for which recombinant virus can be derived and may be from a different AAV serotype than the rAAV genome ITRs, including, but not limited to, AAV serotypes AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV-10 and AAV-11.
  • AAV capsid proteins may be modified to enhance delivery of the recombinant vector. Modifications to capsid proteins are generally known in the art. See, for example, US 20050053922 and US 20090202490, the disclosures of which are incorporated by reference herein in their entirety.
  • a method of generating a packaging cell is to create a cell line that stably expresses all the necessary components for AAV particle production.
  • a plasmid (or multiple plasmids) comprising a rAAV genome lacking AAV rep and cap genes, AAV rep and cap genes separate from the rAAV genome, and a selectable marker, such as a neomycin resistance gene, are integrated into the genome of a cell.
  • AAV genomes have been introduced into bacterial plasmids by procedures such as GC tailing (Samulski et al., 1982, Proc. Natl. Acad. S6.
  • the packaging cell line is then infected with a helper virus such as adenovirus.
  • a helper virus such as adenovirus.
  • packaging cells that produce infectious rAAV.
  • packaging cells may be stably transformed cancer cells such as HeLa cells, 293 cells and PerC.6 cells (a cognate 293 line).
  • packaging cells are cells that are not transformed cancer cells such as low passage 293 cells (human fetal kidney cells transformed with E1 of adenovirus), MRC-5 cells (human fetal fibroblasts), WI-38 cells (human fetal fibroblasts), Vero cells (monkey kidney cells) and FRhL-2 cells (rhesus fetal lung cells).
  • the invention provides rAAV (i.e., infectious encapsidated rAAV particles) comprising a rAAV genome of the invention.
  • the rAAV genome is a self-complementary genome.
  • the genomes of the rAAV lack AAV rep and cap DNA, that is, there is no AAV rep or cap DNA between the ITRs of the genomes.
  • Embodiments include, but are not limited to, the exemplary rAAV including a genome encoding the SOD1 shRNA named “AAV-SOD1-shRNA.”
  • a sequence including the AAV-SOD1-shRNA genome is set out below as an inverted sequence from a plasmid used in production.
  • the rAAV of the invention may be purified by methods standard in the art such as by column chromatography or cesium chloride gradients.
  • Methods for purifying rAAV vectors from helper virus are known in the art and include methods disclosed in, for example, Clark et al., Hum. Gene Ther., 10(6): 1031-1039 (1999); Schenpp and Clark, Methods Mol. Med., 69: 427-443 (2002); U.S. Pat. No. 6,566,118 and WO 98/09657.
  • compositions comprising rAAV of the present invention.
  • Compositions of the invention comprise rAAV in a pharmaceutically acceptable carrier.
  • the compositions may also comprise other ingredients such as diluents and adjuvants.
  • Acceptable carriers, diluents and adjuvants are nontoxic to recipients and are preferably inert at the dosages and concentrations employed, and include buffers such as phosphate, citrate, or other organic acids; antioxidants such as ascorbic acid; low molecular weight polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming
  • Titers of rAAV to be administered in methods of the invention will vary depending, for example, on the particular rAAV, the mode of administration, the treatment goal, the individual, and the cell type(s) being targeted, and may be determined by methods standard in the art. Titers of rAAV may range from about about 1 ⁇ 10 2 , about 1 ⁇ 10 3 , about 1 ⁇ 10 4 , about 1 ⁇ 10 5 , about 1 ⁇ 10 6 , about 1 ⁇ 10 7 , about 1 ⁇ 10 8 , about 1 ⁇ 10 9 , about 1 ⁇ 10 10 , about 1 ⁇ 10 11 , about 1 ⁇ 10 12 , about 1 ⁇ 10 13 to about 1 ⁇ 10 14 or more DNase resistant particles (DRP) per ml.
  • DNase resistant particles DNase resistant particles
  • Dosages may also be expressed in units of viral genomes (vg). Dosages may also vary based on the timing of the administration to a human. These dosages of rAAV may range from about 1 ⁇ 10 4 , about 1 ⁇ 10 5 , about 1 ⁇ 10 6 , about 1 ⁇ 10 7 , about 1 ⁇ 10 8 , about 1 ⁇ 10 9 , about 1 ⁇ 10 10 , about 1 ⁇ 10 11 , about 1 ⁇ 10 12 , about 1 ⁇ 10 13 , about 1 ⁇ 10 14 , about 1 ⁇ 10 15 , about 1 ⁇ 10 16 or more viral genomes per kilogram body weight in an adult.
  • vg viral genomes
  • the dosages of rAAV may range from about about 1 ⁇ 10 4 , about 3 ⁇ 10 4 , about 1 ⁇ 10 5 , about 3 ⁇ 10 5 , about 1 ⁇ 10 6 , about 3 ⁇ 10 6 , about 1 ⁇ 10 7 , about 3 ⁇ 10, about 1 ⁇ 10 8 , about 3 ⁇ 10 8 , about 1 ⁇ 10 9 , about 3 ⁇ 10 9 , about 1 ⁇ 10 10 , about 3 ⁇ 10 10 , about 1 ⁇ 10 11 , about 3 ⁇ 10 11 , about 1 ⁇ 10 12 , about 3 ⁇ 10 12 , about 1 ⁇ 10 13 , about 3 ⁇ 10 13 , about 1 ⁇ 10 14 , about 3 ⁇ 10 14 , about 1 ⁇ 10 15 , about 3 ⁇ 10 15 , about 1 ⁇ 10 16 , about 3 ⁇ 10 16 or more viral genomes per kilogram body weight.
  • compositions comprising rAAV of the present invention.
  • Compositions of the invention comprise rAAV in a pharmaceutically acceptable carrier.
  • the compositions may also comprise other ingredients such as diluents and adjuvants.
  • Acceptable carriers, diluents and adjuvants are nontoxic to recipients and are preferably inert at the dosages and concentrations employed, and include buffers such as phosphate, citrate, or other organic acids; antioxidants such as ascorbic acid; low molecular weight polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming
  • the invention provides methods of transducing a target cell with a rAAV of the invention, in vivo or in vitro.
  • the in vivo methods comprise the step of administering an effective dose, or effective multiple doses, of a composition comprising a rAAV of the invention to a subject, a subject (including a human being), in need thereof. If the dose is administered prior to onset/development of a disorder/disease, the administration is prophylactic. If the dose is administered after the onset/development of a disorder/disease, the administration is therapeutic.
  • an effective dose is a dose that alleviates (eliminates or reduces) at least one symptom associated with the disorder/disease state being treated, that slows or prevents progression to a disorder/disease state, that slows or prevents progression of a disorder/disease state, that diminishes the extent of disease, that results in remission (partial or total) of disease, and/or that prolongs survival.
  • a disease contemplated for treatment with methods of the invention is ALS.
  • “Treatment” thus alleviates (eliminates or reduces) at least one symptom associated with the disorder/disease state being treated (for example, weight loss is eliminated or reduced by at least 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100% or greater), that slows or prevents progression to (onset/development) of a disorder/disease state, that slows or prevents progression of a disorder/disease state, that diminishes the extent of disease, that results in remission (partial or total) of disease, and/or that prolongs survival.
  • a symptom associated with the disorder/disease state being treated for example, weight loss is eliminated or reduced by at least 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%
  • survival is prolonged by at least 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100% or greater.
  • Combination therapies are also contemplated by the invention.
  • Combination as used herein includes both simultaneous treatment or sequential treatments.
  • Combinations of methods of the invention with standard medical treatments e.g., riluzole are specifically contemplated, as are combinations with novel therapies.
  • Administration of an effective dose of the compositions may be by routes standard in the art including, but not limited to, systemic intramuscular, parenteral, intravenous, oral, buccal, nasal, pulmonary, intracranial, intrathecal, intraosseous, intraocular, rectal, or vaginal.
  • Route(s) of administration and serotype(s) of AAV components of the rAAV (in particular, the AAV ITRs and capsid protein) of the invention may be chosen and/or matched by those skilled in the art taking into account the infection and/or disease state being treated and the target cells/tissue(s) that are to express the SOD1 shRNAs.
  • the route of administration is systemic.
  • the route of administration is intrathecal.
  • the route of administration is introcerebroventricular.
  • the route of administration is cisterna magna.
  • the route of administration is by lumbar puncture.
  • Transduction of cells with rAAV of the invention results in sustained expression of SOD1 shRNAs.
  • the present invention thus provides methods of administering/delivering rAAV which express SOD1 shRNA to a subject, preferably a human being.
  • the term “transduction” is used to refer to the administration/delivery of SOD1 shRNAs to a recipient cell either in vivo or in vitro, via a replication-deficient rAAV of the invention resulting in expression of a SOD1 shRNA by the recipient cell.
  • the invention provides methods of administering an effective dose (or doses, administered essentially simultaneously or doses given at intervals) of rAAV that encode SOD1 shRNAs to a subject in need thereof.
  • the invention provides methods of delivering a polynucleotide encoding an shRNA of the invention across the BBB comprising systemically administering a rAAV with a genome including the polynucleotide to a subject.
  • the rAAV genome is a self complementary genome.
  • the rAAV genome is a single-stranded genome.
  • the rAAV is a rAAV9.
  • the rAAV is a rAAV2.
  • the rAAV is a rAAVrh74.
  • the methods systemically deliver polynucleotides across the BBB to the central and/or peripheral nervous system. Accordingly, a method is provided of delivering a polynucleotide to the central nervous system comprising systemically administering a rAAV with a self-complementary genome including the genome to a subject. In some embodiments, the polynucleotide is delivered to brain. In some embodiments, the polynucleotide is delivered to the spinal cord. Also provided is a method of delivering a polynucleotide to the peripheral nervous system comprising systemically administering a rAAV with a self-complementary genome including the polynucleotide to a subject is provided.
  • the polynucleotide is delivered to a lower motor neuron.
  • the rAAV genome is a self complementary genome.
  • the rAAV genome is a single-stranded genome.
  • the rAAV is a rAAV9.
  • the rAAV is a rAAV2.
  • the rAAV is a rAAVrh74.
  • the invention provides methods of delivering a polynucleotide to the central nervous system of a subject in need thereof comprising intrathecal delivery of rAAV with a genome including the polynucleotide.
  • the rAAV genome is a self complementary genome.
  • the rAAV genome is a single-stranded genome.
  • the rAAV is a rAAV9.
  • the rAAV is a rAAV2.
  • the rAAV is a rAAVrh74.
  • a non-ionic, low-osmolar contrast agent is also delivered to the subject, for example, iobitridol, iohexol, iomeprol, iopamidol, iopentol, iopromide, ioversol or ioxilan.
  • Embodiments of the invention employ rAAV to deliver polynucleotides to nerve, glial cells and endothelial cells.
  • the nerve cell is a lower motor neuron and/or an upper motor neuron.
  • the glial cell is a microglial cell, an oligodendrocyte and/or an astrocyte.
  • the rAAV is used to deliver a polynucleotide to a Schwann cell.
  • FIG. 1 AAV9 transduction pattern and persistence in SOD1 G93A mice.
  • Spinal cords were examined for GFP, ChAT (motor neuron marker) and GFAP (astrocyte marker) expression.
  • Temporal vein injection of AAV9-CB-GFP at P1 resulted in efficient transduction of motor neurons and glia in SOD1 G93A mice (a,f,k,p).
  • Tail vein injection at P21 (b,g,l,q) predominantly targeted astrocytes with few GFP positive motor neurons.
  • AAV9-CB-GFP was intravenously injected at P1 and P21 in SOD1 G93A animals that were sacrificed at end stage ( ⁇ P130).
  • Immunofluorescence analysis of lumbar ventral horn (c,d,h,i,m,n,r,s) demonstrated that GFP expression was maintained in astrocytes throughout the disease course.
  • SOD1 mediated inflammation and damage would affect AAV9 transduction, we intravenously injected SOD1 G93A mice at P85 and harvested their spinal cords at endstage. There was no difference observed in the transduction pattern of SOD1 G93A mice treated at P21 or P85.
  • Insets in (r-t) show co-localization between GFP and GFAP signal.
  • AAV adeno-associated virus; P1, postnatal day 1; P21, postnatal day 21; P85, postnatal day 85; GFP, green fluorescent protein; ChAT, choline acetyltransferase; GFAP, glial fibrillary acidic protein.
  • FIG. 2 shRNA constructs show efficient reduction of human SOD1 protein in vitro and in vivo.
  • shRNA 130 was packaged into AAV9 and injected into SOD1 G93A mice at either P1 or P21.
  • FIG. 3 Intravenous delivery of AAV9-SOD1-shRNA improves survival and motor performance in SOD1 G93A mice.
  • FIG. 4 Intravenous injection of AAV9-SOD1-shRNA reduces mutant protein in spinal cords of SOD1 G93A mice.
  • (a-d) Images of lumbar spinal cord sections from uninjected (a), P1 injected (b), P21 injected (c) and P85 injected (d) mice were captured with identical microscope settings to qualitatively show SOD1 levels at end stage. SOD1 levels inversely correlate with survival.
  • FIG. 5 AAV9-SOD1-shRNA improves survival and motor performance in SOD1 G37R mice treated after disease onset.
  • FIG. 6 Intravenous injection of AAV9 in adult SOD1 G37R mice targets astrocytes and motor neurons within the spinal cord.
  • a-h Immunofluorescence analysis revealed neuronal as well as glial transduction in both AAV9-CB-GFP (a-d) and AAV9-SOD1-shRNA treated (e-h) mice.
  • GFP green fluorescent protein
  • ChAT choline acetyltransferase
  • GFAP glial fibrillary acidic protein
  • SOD1 superoxide dismutase 1.
  • FIG. 7 Intrathecal infusion of AAV9-SOD1-shRNA in non-human primates leads to efficient reduction in SOD1 levels.
  • a myelogram shortly after intrathecal infusion of AAV9-SOD1-shRNA mixed with contrast shows proper delivery into the subarachnoid space of a cynomolgus macaque . Arrows show diffusion of the contrast agent along the entire spinal cord.
  • SOD1 Superoxide dismutase 1.
  • FIG. 8 Lumbar intrathecal infusion of AAV9-SOD1-shRNA leads to efficient transduction of motor neurons and non-neuronal cells in the cervical, thoracic and lumbar cord resulting in reduction of SOD1.
  • GFP+/Chat+ cell counts show a caudal to rostral gradient of motor neuron transduction ranging from 85% of transduced cells in the lumbar region to more than 50% in the cervical region.
  • SOD1 mRNA levels in cervical, thoracic and lumbar cord section homogenates analyzed by qRT-PCR show significant reduction in SOD1 transcript, consistently with motor neuron transduction.
  • SOD1 levels were normalized to ⁇ -actin and AAV9-SOD1-shRNA injected animals were compared to an AAV9-CB-GFP injected control.
  • FIG. 9 Design of a clinical SOD1 shRNA construct.
  • Original AAV SOD1 shRNA construct contains shRNA sequence against human SOD1 under H1 promoter followed by the expression cassette for GFP which includes CMV enhancer, CBA promoter, modified SV40 intron, and GFP transgene sequence followed by bGH PolyA terminator.
  • SOD1 shRNA expression cassette and GFP expression cassette are flanked by AAV2 ITRs which ensures the packaging of the complete flanked sequence in AAV9 capsid.
  • the GFP expression cassette is replaced by a stuffer element that contains tandem, noncoding sequences from FDA approved DNA vaccines.
  • ITR inverted terminal repeats
  • shRNA small hairpin RNA
  • SOD1 superoxide dismutase 1
  • CMV cytomegalo virus enhancer
  • CBA Chicken ⁇ -actin promoter
  • GFP green fluorescent protein
  • bGH pA bovine growth hormone poly A terminator.
  • FIG. 10 Schematic of clinical SOD1 shRNA construct. Different restriction sites are placed in the clinical SOD1 shRNA construct that allow the cloning of multiple shRNA expression cassettes while maintaining the total distance between the two ITRs.
  • FIG. 11 In vitro transfection of clinical SOD1 shRNA construct efficiently reduces human SOD1 protein in HEK293 cells. Representative microscopic fields showing bright-field images of non-transfected control (a), AAV SOD1 shRNA transfected (b) and shuttle vector pJet SOD1 shRNA transfected (c,d) HEK 293 cells, 72 hrs post transfection. Corresponding fluorescence images reveal the lack of GFP fluorescence from pJet SOD1 shRNA transfected HEK 293 cells (g,h) as compared to AAV SOD1 shRNA transfected cells (f).
  • FIG. 12 Schematic of cloning strategy for clinical AAV SOD1 shRNA vector.
  • Clinical SOD1 shRNA construct was cloned into AAV CB MCS vector using Kpn1/SPh1 sites.
  • Kpn1/SPh1 double digest of AAV CB MCS plasmid results in the release of the complete transgene expression cassette from this vector which is further replaced with clinical SOD1 shRNA construct carrying SOD1 shRNA expression cassette and stuffer sequence.
  • FIG. 13 Clinical AAV SOD1 shRNA efficiently reduces human SOD1 levels in vitro.
  • HEK293 cells were transfected with clinical AAV SOD1 shRNA plasmid by Calcium phosphate method. Representative microscopic fields showing brightfield images of non-transfected control, AAV SOD1 shRNA and Clinical AAV SOD1 shRNA transfected cells respectively, 72 hrs post-transfection (a-c). Successful removal of GFP from clinical AAV SOD1 shRNA was confirmed by lack of GFP expression in Clinical AAV SOD1 shRNA transfected cells (f,g).
  • FIGS 1 AAV9-shRNA-SOD1 administration is well tolerated in WT mice.
  • Female and male WT animals were injected with AAV9-SOD1-shRNA at P1 or P21 and monitored up to 6 months of age.
  • (a,b) Both male and female treated mice showed steady increase in body mass as compared to control animals.
  • (c,d) Rotarod performance and (e,f) hind limb grip strength were not affected by P1 or P21 treatment in both groups as compared to respective controls.
  • n 5 per group.
  • WT wild type; P1, postnatal day 1; P21, postnatal day 21.
  • FIGS 2 Hematology and Serum Chemistry of AAV9-SOD1-shRNA treated WT animals.
  • a-m Blood was collected from P1 (green) or P21 (red) treated and control (gray) WT animals at 150 days of age for hematology studies. No significant differences were observed between treated and control animals.
  • FIG. 1 u a diagram illustrating the percent astrocyte transduction achieved in these mice.
  • FIG. 4 Intravenous injection of AAV9-SOD1-shRNA efficiently reduces levels of mutant SOD1 protein in spinal cords of SOD1 G37R mice.
  • AAV9-CB-GFP or AAV9-SOD1-shRNA was injected in SOD1 G37R mice and spinal cords were harvested at end stage and analyzed by western blot for human SOD1 protein levels.
  • FIG. 5 shRNA 130 efficiently reduces the levels of monkey SOD1 in vitro.
  • Transduction efficiency was high in SOD1 G93A astrocytes with GFP expressed in 34 ⁇ 2% and 54 ⁇ 3%, respectively, of P1 and P21 injected spinal grey matter astrocytes (defined by immunoreactivity for GFAP). This efficiency was similar to our previous report of 64 ⁇ 1% in P21 injected wild type animals 18 . Motor neurons were a prominent cell type transduced at all levels of the spinal cords of P1 injected SOD1 G93A animals (62 ⁇ 1%), compared with significantly lower targeting to motor neurons in P21 injected animals (8 ⁇ 1%).
  • SOD1 mutant mediated damage including astrocytic and microglial activation and early changes in the blood brain barrier develop during disease in mice in SOD1 mutant mice 20
  • this damage affected AAV9 transduction.
  • Analysis of the spinal cords revealed that the transduction pattern seen in P85 animals was similar to P21 treated animals with astrocytes as the predominant cell type transduced at all levels (51 ⁇ 6% GFP+/GFAP+ cells in lumbar grey matter).
  • shRNA constructs targeting human SOD1 were generated and obtained from the Life Technologies design tool.
  • the constructs that had a minimum of four base mismatches compared to the mouse mRNA sequence correspond to record number CCDS33536.1 in the NCBI CCDS database.
  • These constructs were cloned in pSilencer 3.1 (Genscript) under the human H1 promoter and tested in vitro.
  • shRNA 130 along with H1 promoter was further cloned into an AAV vector along with a reporter GFP under Chicken Beta-Actin promoter to identify the transduced cells. Human 293 cells were transfected with each cassette.
  • the HEK-293 cells were maintained in IMDM medium containing 10% FBS, 1% L-glutamine and 1% penicillin/streptomycin. Upon reaching ⁇ 60% confluence, cells were transfected with pSilencer 3.1 containing the shRNAs being tested. Protein lysates were prepared 72 hours post transfection and analyzed for SOD1 levels by western blot. All four sequences reduced SOD1 protein levels by >50% ( FIGS. 2 b,c ).
  • shRNA130 was selected for further experiments because it produced the most consistent knockdown across three separate transfection experiments. It was cloned into a self-complementary AAV9 vector that also contained a GFP gene whose expression would identify transduced cells (referred to as AAV9-SOD1-shRNA). Self-complementary AAV9-SOD1-shRNA was produced by transient transfection procedures using a double-stranded AAV2-ITR-based CB-GFP vector, with a plasmid encoding Rep2Cap9 sequence as previously described along with an adenoviral helper plasmid pHelper (Stratagene, Santa Clara, Calif.) in 293 cells 18 .
  • AAV9-SOD1-shRNA self-complementary AAV9 vector that also contained a GFP gene whose expression would identify transduced cells
  • AAV9-SOD1-shRNA For neonatal mouse injections, postnatal day 1-2 SOD1 G93A pups were utilized. Total volume of 50 ⁇ l containing 5 ⁇ 10 11 DNAse resistant viral particles of AAV9-SOD1-shRNA (Virapur LLC, San Diego, Calif.) was injected via temporal vein as previously described 18 . A correct injection was verified by noting blanching of the vein. After the injection, pups were returned to their cage.
  • AAV9-SOD1-shRNA is Safe and Well Tolerated in Wild Type Mice
  • animals were placed in a restraint that positioned the mouse tail in a lighted, heated groove. The tail was swabbed with alcohol then injected intravenously with AAV9-SOD1-shRNA.
  • loxSOD1 G37R ALS mice carrying a human mutant SOD1 G37R transgene flanked by lox p sites under its endogenous promoter, were maintained in as previously described 37 .
  • FIG. 6 Histological examination of end-stage SOD1 G37R treated animals revealed similar levels of intraspinal cell transduction in animals treated with AAV9-SOD1-shRNA or AAV9-GFP ( FIG. 6 ). GFP expression was predominantly observed within motor neurons and astrocytes of both groups, and SOD1 expression was detectably decreased only in animals that received AAV9-SOD1-shRNA ( FIGS. 6 k,o ). Immunoblotting of whole spinal cord extracts from end stage SOD1 G37R mice revealed an 80% reduction in hSOD1 protein levels in AAV9-SOD1-shRNA treated animals compared to controls ( Figure S4 ).
  • AAV9 was injected intrathecally via lumbar puncture. This method was chosen over systemic delivery to decrease the amount of virus required and to minimize any effects from reduction of SOD1 in peripheral tissues.
  • One year old cynomolgus macaques ( Macaca fascicularis ) with average body weight of 2 kg were used for this study at the Mannheimer Foundation. Regular monitoring of overall health and body weight was performed prior and after the injections to assess the welfare of the animals.
  • cDNA was prepared using RT 2 First strand synthesis kit (SABiosciences). SOD1 transcript levels were analyzed by qRT-PCR which revealed that the monkey SOD1 mRNA was reduced by ⁇ 75% in 130 shRNA transduced cells compared to mock transduced control cells ( Figure S 5 ).
  • the AAV9-SOD1-shRNA virus (1 ⁇ 10 13 vg/kg) was infused along with contrast agent via lumbar puncture into the subarachnoid space of three male cynomolgus macaques and one control subject was injected with AAV9-CB-GFP (1 ⁇ 10 13 vg/kg) ( FIG. 7 a ).
  • Each intrathecal injection was performed by lumbar puncture into the subarachnoid space of the lumbar thecal sac.
  • AAV9 was resuspended with omnipaque (iohexol), an iodinated compound routinely used in the clinical setting.
  • Iohexol is used to validate successful subarachnoid space cannulation and was administered at a dose of 100 mg/Kg.
  • the subject was placed in the lateral decubitus position and the posterior midline injection site at ⁇ L4/5 level identified (below the conus of the spinal cord). Under sterile conditions, a spinal needle with stylet was inserted and subarachnoid cannulation was confirmed with the flow of clear CSF from the needle.
  • 0.8 ml of CSF was drained, immediately followed by injection with a mixture containing 0.7 mL iohexol (300 mg/ml formulation) mixed with 2.1 mL of virus (2.8 ml total).
  • FIGS. 8 a - c Motor neuron counts revealed a caudal to rostral gradient in cell transduction, with the cervical region showing more than 50% of GFP/Chat+ motor neurons, increasing to 65% in the thoracic region and reaching a remarkable 80% in the lumbar region ( FIG. 8 d ).
  • qRT-PCR for SOD1 was performed on whole section homogenates from cervical, thoracic and lumbar cord segments.
  • mice Control and treated SOD1 G93A mice were sacrificed at either 21 days post injection or at endstage for immunohistochemical analysis. Animals were anesthetized with xylazene/ketamine cocktail, transcardially perfused with 0.9% saline, followed by 4% paraformaldehyde. Spinal cords were harvested, cut into blocks of tissue 5-6 mm in length, and then cut into 40 ⁇ m thick transverse sections on a vibratome (Leica, Bannockburn, Ill.). Serial sections were kept in a 96-well plate that contained 4% paraformaldehyde and were stored at 4° C.
  • End stage loxSOD1 G37R mice were anesthetized using isoflurane and perfused with 4% paraformaldehyde.
  • Spinal cord segments including cervical, thoracic and lumbar segments were dissected.
  • spinal cords were frozen in isopentane at ⁇ 65° C., and serial 30 ⁇ m coronal sections were collected free floating using sliding microtome.
  • mice were sacrificed at 180 days of age. Animals were anesthetized using xylazene/ketamine cocktail and perfused with 0.9% saline. Different tissues were removed and stored in 10% buffered formalin. These tissues were further processed, blocked and mounted for hematoxilin & eosin staining by the Nationalwide Children's Hospital Morphology Core.
  • Cynomolgus monkeys injected with virus were euthanized 2 weeks post injection. Animals were anesthetized with sodium pentobarbital at the dose of 80-100 mg/kg intravenously and perfused with saline solution. Brain and spinal cord dissection were performed immediately and tissues were processed either for nucleic acid isolation (snap frozen) or post-fixed in 4% paraformaldehyde and subsequently cryoprotected with 30% sucrose and frozen in isopentane at ⁇ 65° C. 12 ⁇ m coronal sections were collected from lumbar cord using a cryostat for free floating immunostaining.
  • Primary antibodies used were as follows: rabbit anti-GFP (1:400, Invitrogen, Carlsbad, Calif.), rabbit anti-SOD1 (1:200, Cell signaling, Danvers, Mass.), goat anti-ChAT (1:50 Millipore, Billerica, Mass.), mouse anti-GFAP (1:200, Millipore, Billerica, Mass.), chicken anti GFAP (1:400, Abcam, Cambridge, Mass.), and rabbit anti-Ibal (1:400, Wako, Richmond Va.). Tissues were incubated in primary antibody at 4° C. for 48-72 hours then washed three times with TBS.
  • tissue were incubated for 2 hours at room temperature in the appropriate FITC-, Cy3-, or Cy5-conjugated secondary antibodies (1:200 Jackson Immunoresearch, Westgrove, Pa.) and DAPI (1:1000, Invitrogen, Carlsbad, Calif.). Tissues were then washed three times with TBS, mounted onto slides then coverslipped with PVA-DABCO. All images were captured on a Zeiss-laser-scanning confocal microscope.
  • monkey spinal cord sections were washed three times in TBS, blocked for 2 h at RT in 10% donkey serum and 1% Triton X-100. Sections were then incubated overnight at 4° C. with rabbit anti-GFP primary antibody (1:1000 Invitrogen, Carlsbad, Calif.) diluted in blocking buffer. The following day, tissues were washed with TBS 3 times, incubated with biotinylated secondary antibody anti-rabbit (1:200 Jackson Immunoresearch, Westgrove, Pa.) in blocking buffer for 30 min at RT, washed 3 times in TBS and incubated for 30 min at RT with ABC (Vector, Burlingame, Calif.).
  • Sections were then washed for 3 times in TBS and incubated for 2 min with DAB solution at RT and washed with distilled water. These were then mounted onto slides and covered with coverslips in mounting medium. All images were captured with the Zeiss Axioscope.
  • Spinal cords were harvested from P1, P21 injected and control SOD1 G93A mice 21 days post injection and from treated and control monkeys 2 weeks post injection of AAV9-SOD1-shRNA.
  • Spinal cords were homogenized and protein lysates were prepared using T-Per (Pierce) with protease inhibitor cocktail. Samples were resolved on SDS-PAGE according to manufacturer's instructions.
  • RNA from laser captured cells or whole spinal cord sections from the cervical, thoracic and lumbar segments was isolated using the RNaqueous Micro Kit (Ambion, Grand Island, N.Y.) according to manufacturer's instructions. RNA was then reverse-transcribed into cDNA using the RT 2 HT First Strand Kit (SABiosciences, Valencia, Calif.). 12.5 ng RNA were used in each Q-PCR reaction using SyBR Green (Invitrogen, Carlsbad, Calif.) to establish the relative quantity of endogenous monkey SOD1 transcript in animals who had received the AAV9-SOD1-shRNA compared to animals who had received only AAV9-GFP. Each sample was run in triplicate and relative concentration calculated using the ddCt values normalized to endogenous actin transcript.
  • SOD1 G93A mice were monitored for changes in body mass twice a week.
  • loxSOD1 G37R mice were weighed on a weekly basis. Motor coordination was recorded using a rotarod instrument (Columbus Instruments, Columbus, Ohio). Each weekly session consisted of three trials on the accelerating rotarod beginning at 5 rpm/min. The time each mouse remained on the rod was registered.
  • Both SOD1 G93A and loxSOD1 G37R mice were subjected to weekly assessment of hindlimb grip strength using a grip strength meter (Columbus Instruments, Columbus, Ohio). Each weekly session consisted of 3 (SOD1 G93A mice) or 5 (loxSOD1 G37R mice) tests per animal.
  • End stage was defined as an artificial death point when animals could no longer “right” themselves within 30 sec after being placed on its back.
  • Onset and disease progression were determined from retrospective analysis of the data.
  • Disease onset is defined as the age at which the animal reached its peak weight.
  • Disease duration is defined as the time period between disease onset and end stage.
  • Early disease duration is the period between peak weight and loss of 10% of body weight while late disease duration is defined as the period between 10% loss of body weight until disease end stage. Due to shorter life span of SOD1 G93A animals, we did not assess the distinction between the early and late progression.
  • mice For toxicity analysis following injection at P1 or P21, treated and control WT mice were subjected to behavioral analysis starting at ⁇ 30 days of age and monitored up to 6 months. Body mass was recorded weekly while rotarod performance and hindlimb grip strength were recorded biweekly.
  • Blood samples were collected in (K2) EDTA microtainer tubes (BD) from treated and control WT mice at 150 days of age by mandibular vein puncture. The same animals were bled at 180 days of age and blood was collected in serum separator microtainer tubes. The blood was allowed to clot for an hour and was then centrifuged at 10,000 rpm for 5 minutes. The clear upper phase (serum) was collected and frozen at ⁇ 80° C. Hematological and serum analysis were conducted by Ani Lytics Inc, Gaithersburg, Md.
  • the AAV SOD1 shRNA vector described in Example 2 carries shRNA against human SOD1 sequence under the H1 promoter ( FIG. 9 a ).
  • the same vector also contains a GFP expression cassette which expresses GFP under a CBA promoter.
  • the other regulatory elements present in this cassette include CMV enhancer, SV40 intron and bGH PolyA terminator sequence.
  • the SOD1 shRNA construct of Example 2 was modified by replacing the GFP expression cassette with a non-coding stuffer sequence while maintaining the size of the total DNA construct flanked by the ITRs ( FIG. 9 b ). This is important as the distance between the two ITR sequences greatly affects the packaging capacity of the flanked construct into AAV9 capsids[321-324].
  • Clinical SOD1 shRNA construct has shRNA against human SOD1 under H1 promoter which is followed by the non-coding stuffer sequence.
  • This construct is designed in such a way that multiple shRNA expression cassettes can be added to the final vector by simultaneous removal of the stuffer sequence. Restriction endonuclease sites have been added to the stuffer sequence so that a part of the stuffer can be removed when another shRNA expression cassette is added ( FIG. 10 ). This simultaneous removal and addition of DNA sequences would help maintaining the optimal size of the whole construct between the ITRs ( ⁇ 2.0 kb) to achieve efficient packaging.
  • HEK293 cells were transfected with pJet SOD1 shRNA plasmid using Calcium Phosphate method.
  • AAV SOD1 shRNA plasmid was used as a positive control.
  • Immunofluorescence analysis of HEK293 cells, 72 hrs post transfection revealed the lack of native GFP fluorescence from pJet SOD1 shRNA transfected cells as compared to AAV9 SOD1 shRNA transfected cells. Immunoblot analysis of cell lysates from these cells further confirmed the successful replacement of GFP from pJet SOD1 shRNA plasmid.
  • pJet SOD1 shRNA resulted in efficient downregulation of SOD1 protein levels (>50%), similar to AAV SOD1 shRNA plasmid. See FIG. 11 .
  • Clinical SOD1 shRNA construct was further cloned into an AAV.CB.MCS vector using Kpn1/Sph1 sites to generate clinical AAV SOD1 shRNA plasmid ( FIG. 12 ).
  • AAV.CB.MCS was generated from AAV.CB.GFP plasmid obtained from merion Scientific by replacing GFP with a multiple cloning site (MCS).
  • Cloning of clinical SOD1 shRNA construct at Kpn1/Sph1 sites puts it between the two AAV2 ITRs which facilitates the packaging of the construct in AAV9 viral capsids. See FIG. 12 .
  • Clinical AAV SOD1 shRNA plasmid was screened with restriction endonucleases to confirm the presence of SOD1 shRNA expression cassette (Xba1 digest), stuffer sequence (EcoRV/Pme1 double digest) and also intact ITR sequences (Sma1 digest).
  • Clinical AAV SOD1 shRNA plasmid was transfected in HEK293 cells to determine its knockdown efficiency. Similar to the pJet SOD1 shRNA plasmid, clinical AAV SOD1 shRNA transfected cells were devoid of any GFP expression as evident by immunofluorescence ( FIG. 13 a - f ) and immunoblot assay ( FIG. 13 g ). More importantly, clinical AAV SOD1 shRNA efficiently reduced human SOD1 protein levels in HEK293 cells by more than 50% ( FIG. 13 g,h ). Altogether, these results confirmed the successful generation of clinical AAV SOD1 shRNA vector with functional SOD1 shRNA expression cassette and complete removal of the transgene expression cassette.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Immunology (AREA)
  • Dermatology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US14/914,861 2013-08-27 2014-08-26 Products and methods for treatment of familial amyotrophic lateral sclerosis Abandoned US20160272976A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/914,861 US20160272976A1 (en) 2013-08-27 2014-08-26 Products and methods for treatment of familial amyotrophic lateral sclerosis

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361870585P 2013-08-27 2013-08-27
US14/914,861 US20160272976A1 (en) 2013-08-27 2014-08-26 Products and methods for treatment of familial amyotrophic lateral sclerosis
PCT/US2014/052753 WO2015031392A1 (en) 2013-08-27 2014-08-26 Products and methods for treatment of amyotrophic lateral sclerosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/052753 A-371-Of-International WO2015031392A1 (en) 2013-08-27 2014-08-26 Products and methods for treatment of amyotrophic lateral sclerosis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/041,381 Continuation US10793861B2 (en) 2013-08-27 2018-07-20 Products and methods for treatment of familial amyotrophic lateral sclerosis

Publications (1)

Publication Number Publication Date
US20160272976A1 true US20160272976A1 (en) 2016-09-22

Family

ID=51494538

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/914,861 Abandoned US20160272976A1 (en) 2013-08-27 2014-08-26 Products and methods for treatment of familial amyotrophic lateral sclerosis
US16/041,381 Active 2034-11-16 US10793861B2 (en) 2013-08-27 2018-07-20 Products and methods for treatment of familial amyotrophic lateral sclerosis
US16/997,304 Abandoned US20210108209A1 (en) 2013-08-27 2020-08-19 Products and Methods for Treatment of Familial Amyotrophic Lateral Sclerosis

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/041,381 Active 2034-11-16 US10793861B2 (en) 2013-08-27 2018-07-20 Products and methods for treatment of familial amyotrophic lateral sclerosis
US16/997,304 Abandoned US20210108209A1 (en) 2013-08-27 2020-08-19 Products and Methods for Treatment of Familial Amyotrophic Lateral Sclerosis

Country Status (15)

Country Link
US (3) US20160272976A1 (da)
EP (3) EP4219727A3 (da)
CA (1) CA2922500A1 (da)
CY (1) CY1123272T1 (da)
DK (2) DK3702466T3 (da)
ES (2) ES2936220T3 (da)
FI (1) FI3702466T3 (da)
HR (2) HRP20230015T1 (da)
HU (2) HUE050546T2 (da)
LT (2) LT3039146T (da)
PL (1) PL3702466T3 (da)
PT (2) PT3702466T (da)
RS (2) RS60434B1 (da)
SI (2) SI3039146T1 (da)
WO (1) WO2015031392A1 (da)

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160120960A1 (en) * 2013-05-15 2016-05-05 Regents Of The University Of Minnesota Adeno-associated virus mediated gene transfer to the central nervous system
US10280418B2 (en) * 2014-03-18 2019-05-07 Univeristy Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
WO2019094253A1 (en) 2017-11-08 2019-05-16 Avexis Inc. Means and method for preparing viral vectors and uses of same
US10300146B2 (en) 2008-05-28 2019-05-28 University Of Massachusetts Isolation of novel AAV's and uses thereof
US10370432B2 (en) 2014-10-03 2019-08-06 University Of Massachusetts Heterologous targeting peptide grafted AAVS
US10457940B2 (en) 2016-09-22 2019-10-29 University Of Massachusetts AAV treatment of Huntington's disease
US10480011B2 (en) 2014-10-21 2019-11-19 University Of Massachusetts Recombinant AAV variants and uses thereof
US10570395B2 (en) 2014-11-14 2020-02-25 Voyager Therapeutics, Inc. Modulatory polynucleotides
US10584337B2 (en) 2016-05-18 2020-03-10 Voyager Therapeutics, Inc. Modulatory polynucleotides
US10584321B2 (en) 2015-02-13 2020-03-10 University Of Massachusetts Compositions and methods for transient delivery of nucleases
US10597660B2 (en) 2014-11-14 2020-03-24 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
US10689420B2 (en) 2009-05-28 2020-06-23 University Of Massachusetts AAV's and uses thereof
US10689653B2 (en) 2014-06-03 2020-06-23 University Of Massachusetts Compositions and methods for modulating dysferlin expression
US10731178B2 (en) 2010-04-23 2020-08-04 University Of Massachusetts CNS targeting AAV vectors and methods of use thereof
US10894949B2 (en) 2014-02-19 2021-01-19 University Of Massachusetts Recombinant AAVS having useful transcytosis properties
US11046955B2 (en) 2015-04-24 2021-06-29 University Of Massachusetts Modified AAV constructs and uses thereof
US11060088B2 (en) 2016-02-12 2021-07-13 University Of Massachusetts Anti-angiogenic miRNA therapeutics for inhibiting corneal neovascularization
US11207426B2 (en) 2016-04-05 2021-12-28 University Of Massachusetts Compositions and methods for selective inhibition of grainyhead-like protein expression
US11253576B2 (en) 2015-10-22 2022-02-22 University Of Massachusetts Methods and compositions for treating metabolic imbalance in neurodegenerative disease
US11253612B2 (en) 2016-04-15 2022-02-22 The Trustees Of The University Of Pennsylvania Gene therapy for treating mucopolysaccharidosis type II
US11413356B2 (en) 2016-04-15 2022-08-16 University Of Massachusetts Methods and compositions for treating metabolic imbalance
US11426469B2 (en) 2015-10-22 2022-08-30 University Of Massachusetts Prostate-targeting adeno-associated virus serotype vectors
US11434502B2 (en) 2017-10-16 2022-09-06 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (ALS)
US11578340B2 (en) 2016-10-13 2023-02-14 University Of Massachusetts AAV capsid designs
US11603542B2 (en) 2017-05-05 2023-03-14 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
WO2023073526A1 (en) 2021-10-25 2023-05-04 Novartis Ag Methods for improving adeno-associated virus (aav) delivery
US11739330B2 (en) 2017-09-22 2023-08-29 University Of Massachusetts SOD1 dual expression vectors and uses thereof
US11819539B2 (en) 2017-09-22 2023-11-21 The Trustees Of The University Of Pennsylvania Gene therapy for treating Mucopolysaccharidosis type II
US11826433B2 (en) 2016-02-02 2023-11-28 University Of Massachusetts Method to enhance the efficiency of systemic AAV gene delivery to the central nervous system
US11859179B2 (en) 2017-05-09 2024-01-02 University Of Massachusetts Methods of treating amyotrophic lateral sclerosis (ALS)
US11882815B2 (en) 2016-06-15 2024-01-30 University Of Massachusetts Recombinant adeno-associated viruses for delivering gene editing molecules to embryonic cells
WO2024015794A3 (en) * 2022-07-12 2024-03-07 The Research Institute At Nationwide Children's Hospital Adeno-associated virus gene therapy products and methods
US11931375B2 (en) 2017-10-16 2024-03-19 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (ALS)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10131904B2 (en) 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
EP3336188B1 (en) 2008-09-22 2020-05-06 Phio Pharmaceuticals Corp. Reduced size self-delivering rnai compounds
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
EP2699680B1 (en) 2011-04-21 2017-12-27 University of Massachusetts Raav-based compositions and methods for treating alpha-1 anti-trypsin deficiencies
DK3702466T3 (da) * 2013-08-27 2023-03-20 Res Inst Nationwide Childrens Hospital Produkter og fremgangsmåder til behandling af amyotrofisk lateral sclerose
US9725719B2 (en) 2013-11-05 2017-08-08 The Research Institute At Nationwide Children's Hospital Compositions and methods for inhibiting NF-κB and SOD-1 to treat amyotrophic lateral sclerosis
AU2015240761B2 (en) 2014-04-01 2019-09-12 Biogen Ma Inc. Compositions for modulating SOD-1 expression
WO2017007825A1 (en) * 2015-07-06 2017-01-12 Rxi Pharmaceuticals Corporation Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
US11001845B2 (en) 2015-07-06 2021-05-11 Phio Pharmaceuticals Corp. Nucleic acid molecules targeting superoxide dismutase 1 (SOD1)
EP3320101B1 (en) 2015-07-07 2021-08-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for expressing a polynucleotide of interest in the peripheral nervous system of a subject
EP3121284A1 (en) 2015-07-22 2017-01-25 Ecole Polytechnique Federale de Lausanne (EPFL) Bicistronic aav vector for rna interference in als
EP3500278B1 (en) 2016-08-19 2024-05-22 University of Florida Research Foundation, Incorporated Compositions for treating conditions using recombinant self-complementary adeno-associated virus
CA3046347A1 (en) 2016-12-07 2018-06-14 University Of Florida Research Foundation, Incorporated Il-1ra cdnas
SG11201909136PA (en) * 2017-03-31 2019-10-30 Staidson Beijing Biopharmaceuticals Co Ltd Shrna expression cassette, polynucleotide sequence carrying the same, and use thereof

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5448517A (en) 1987-06-29 1995-09-05 Kabushiki Kaisha Toshiba Electrically programmable nonvolatile semiconductor memory device with NAND cell structure
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
WO1995013392A1 (en) 1993-11-09 1995-05-18 Medical College Of Ohio Stable cell lines capable of expressing the adeno-associated virus replication gene
AU688428B2 (en) 1993-11-09 1998-03-12 Johns Hopkins University, The Generation of high titers of recombinant AAV vectors
US5658785A (en) 1994-06-06 1997-08-19 Children's Hospital, Inc. Adeno-associated virus materials and methods
US5856152A (en) 1994-10-28 1999-01-05 The Trustees Of The University Of Pennsylvania Hybrid adenovirus-AAV vector and methods of use therefor
JPH10511264A (ja) 1994-12-06 1998-11-04 ターゲティッド ジェネティックス コーポレイション 高力価組換えaavベクターの生成のためのパッケージング細胞株
FR2737730B1 (fr) 1995-08-10 1997-09-05 Pasteur Merieux Serums Vacc Procede de purification de virus par chromatographie
JPH11511326A (ja) 1995-08-30 1999-10-05 ジエンザイム コーポレイション アデノウィルスおよびaavの精製
EP0850313B8 (en) 1995-09-08 2009-07-29 Genzyme Corporation Improved aav vectors for gene therapy
US5910434A (en) 1995-12-15 1999-06-08 Systemix, Inc. Method for obtaining retroviral packaging cell lines producing high transducing efficiency retroviral supernatant
DE69739860D1 (de) 1996-09-06 2010-06-02 Univ Pennsylvania Rekombinante AAV zur Herstellung eines Medikaments für die Gentherapie von Muskelzellen
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
DK1944362T3 (da) 1997-09-05 2016-01-25 Genzyme Corp Fremgangsmåder til fremstilling af hjælpevirusfri præparater med høj titer af rekombinante AAV-vektorer
US6258595B1 (en) 1999-03-18 2001-07-10 The Trustees Of The University Of Pennsylvania Compositions and methods for helper-free production of recombinant adeno-associated viruses
CA2406743A1 (en) 2000-04-28 2001-11-08 The Trustees Of The University Of Pennsylvania Recombinant aav vectors with aav5 capsids and aav5 vectors pseudotyped in heterologous capsids
US9441244B2 (en) 2003-06-30 2016-09-13 The Regents Of The University Of California Mutant adeno-associated virus virions and methods of use thereof
US9233131B2 (en) 2003-06-30 2016-01-12 The Regents Of The University Of California Mutant adeno-associated virus virions and methods of use thereof
WO2005096781A2 (en) * 2004-04-06 2005-10-20 University Of Massachusetts Methods and compositions for treating gain-of-function disorders using rna interference
US20060229268A1 (en) * 2004-12-16 2006-10-12 Alsgen, Llc Small interference RNA (siRNA) molecules for modulating superoxide dismutase (SOD)
EP2019143A1 (en) 2007-07-23 2009-01-28 Genethon CNS gene delivery using peripheral administration of AAV vectors
EP2058401A1 (en) 2007-10-05 2009-05-13 Genethon Widespread gene delivery to motor neurons using peripheral injection of AAV vectors
US10131904B2 (en) * 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
WO2010071832A1 (en) 2008-12-19 2010-06-24 Nationwide Children's Hospital Delivery of polynucleotides across the blood brain barrier using recombinant aav9
EP2826860B1 (en) 2010-04-23 2018-08-22 University of Massachusetts CNS targeting AAV vectors and methods of use thereof
WO2012058220A2 (en) * 2010-10-26 2012-05-03 University Of Massachusetts Anti-sod1 antibodies and uses thereof
WO2013078316A1 (en) 2011-11-23 2013-05-30 Nationwide Children's Hospital, Inc. Recombinant adeno-associated virus delivery of alpha-sarcoglycan polynucleotides
DE102012007232B4 (de) 2012-04-07 2014-03-13 Susanne Weller Verfahren zur Herstellung von rotierenden elektrischen Maschinen
RU2019143431A (ru) * 2012-11-01 2020-04-28 Фэктор Байосайенс Инк. Способы и продукты для экспрессии белков в клетках
DK3702466T3 (da) * 2013-08-27 2023-03-20 Res Inst Nationwide Childrens Hospital Produkter og fremgangsmåder til behandling af amyotrofisk lateral sclerose
JP2015092462A (ja) 2013-09-30 2015-05-14 Tdk株式会社 正極及びそれを用いたリチウムイオン二次電池
JP6202701B2 (ja) 2014-03-21 2017-09-27 株式会社日立国際電気 基板処理装置、半導体装置の製造方法及びプログラム
JP6197169B2 (ja) 2014-09-29 2017-09-20 東芝メモリ株式会社 半導体装置の製造方法

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10905776B2 (en) 2008-05-28 2021-02-02 University Of Massachusetts Isolation of novel AAV's and uses thereof
US10300146B2 (en) 2008-05-28 2019-05-28 University Of Massachusetts Isolation of novel AAV's and uses thereof
US11826434B2 (en) 2008-05-28 2023-11-28 University Of Massachusetts Isolation of novel AAV's and uses thereof
US11834474B2 (en) 2009-05-28 2023-12-05 University Of Massachusetts AAV's and uses thereof
US10689420B2 (en) 2009-05-28 2020-06-23 University Of Massachusetts AAV's and uses thereof
US10731178B2 (en) 2010-04-23 2020-08-04 University Of Massachusetts CNS targeting AAV vectors and methods of use thereof
US20160120960A1 (en) * 2013-05-15 2016-05-05 Regents Of The University Of Minnesota Adeno-associated virus mediated gene transfer to the central nervous system
US9827295B2 (en) * 2013-05-15 2017-11-28 Regents Of The University Of Minnesota Methods to treat mucopolysaccharide type I or deficiency in alpha-L-iduronidase using a recombinant adeno-associated virus encoding alpha-L-iduronidase
US10894949B2 (en) 2014-02-19 2021-01-19 University Of Massachusetts Recombinant AAVS having useful transcytosis properties
US10851375B2 (en) 2014-03-18 2020-12-01 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10954518B2 (en) 2014-03-18 2021-03-23 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US11760999B2 (en) 2014-03-18 2023-09-19 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10711274B2 (en) 2014-03-18 2020-07-14 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10280418B2 (en) * 2014-03-18 2019-05-07 Univeristy Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10689653B2 (en) 2014-06-03 2020-06-23 University Of Massachusetts Compositions and methods for modulating dysferlin expression
US11827886B2 (en) 2014-06-03 2023-11-28 University Of Massachusetts Compositions and methods for modulating dysferlin expression
US10370432B2 (en) 2014-10-03 2019-08-06 University Of Massachusetts Heterologous targeting peptide grafted AAVS
US11014976B2 (en) 2014-10-03 2021-05-25 University Of Massachusetts Heterologous targeting peptide grafted AAVS
US10480011B2 (en) 2014-10-21 2019-11-19 University Of Massachusetts Recombinant AAV variants and uses thereof
US11542525B2 (en) 2014-10-21 2023-01-03 University Of Massachusetts Recombinant AAV variants and uses thereof
US10570395B2 (en) 2014-11-14 2020-02-25 Voyager Therapeutics, Inc. Modulatory polynucleotides
US10920227B2 (en) 2014-11-14 2021-02-16 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
US10597660B2 (en) 2014-11-14 2020-03-24 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
US11198873B2 (en) 2014-11-14 2021-12-14 Voyager Therapeutics, Inc. Modulatory polynucleotides
US11542506B2 (en) 2014-11-14 2023-01-03 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
US10584321B2 (en) 2015-02-13 2020-03-10 University Of Massachusetts Compositions and methods for transient delivery of nucleases
US11920168B2 (en) 2015-02-13 2024-03-05 University Of Massachusetts Compositions and methods for transient delivery of nucleases
US11046955B2 (en) 2015-04-24 2021-06-29 University Of Massachusetts Modified AAV constructs and uses thereof
US11253576B2 (en) 2015-10-22 2022-02-22 University Of Massachusetts Methods and compositions for treating metabolic imbalance in neurodegenerative disease
US11426469B2 (en) 2015-10-22 2022-08-30 University Of Massachusetts Prostate-targeting adeno-associated virus serotype vectors
US11826433B2 (en) 2016-02-02 2023-11-28 University Of Massachusetts Method to enhance the efficiency of systemic AAV gene delivery to the central nervous system
US11060088B2 (en) 2016-02-12 2021-07-13 University Of Massachusetts Anti-angiogenic miRNA therapeutics for inhibiting corneal neovascularization
US11851657B2 (en) 2016-02-12 2023-12-26 University Of Massachusetts Anti-angiogenic miRNA therapeutics for inhibiting corneal neovascularization
US11207426B2 (en) 2016-04-05 2021-12-28 University Of Massachusetts Compositions and methods for selective inhibition of grainyhead-like protein expression
US11413356B2 (en) 2016-04-15 2022-08-16 University Of Massachusetts Methods and compositions for treating metabolic imbalance
US11253612B2 (en) 2016-04-15 2022-02-22 The Trustees Of The University Of Pennsylvania Gene therapy for treating mucopolysaccharidosis type II
US10584337B2 (en) 2016-05-18 2020-03-10 Voyager Therapeutics, Inc. Modulatory polynucleotides
US11193129B2 (en) 2016-05-18 2021-12-07 Voyager Therapeutics, Inc. Modulatory polynucleotides
US11882815B2 (en) 2016-06-15 2024-01-30 University Of Massachusetts Recombinant adeno-associated viruses for delivering gene editing molecules to embryonic cells
US10457940B2 (en) 2016-09-22 2019-10-29 University Of Massachusetts AAV treatment of Huntington's disease
US11773392B2 (en) 2016-09-22 2023-10-03 University Of Massachusetts AAV treatment of Huntington's disease
US11046957B2 (en) 2016-09-22 2021-06-29 University Of Massachusetts AAV treatment of Huntington's disease
US11578340B2 (en) 2016-10-13 2023-02-14 University Of Massachusetts AAV capsid designs
US11603542B2 (en) 2017-05-05 2023-03-14 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
US11859179B2 (en) 2017-05-09 2024-01-02 University Of Massachusetts Methods of treating amyotrophic lateral sclerosis (ALS)
US11819539B2 (en) 2017-09-22 2023-11-21 The Trustees Of The University Of Pennsylvania Gene therapy for treating Mucopolysaccharidosis type II
US11739330B2 (en) 2017-09-22 2023-08-29 University Of Massachusetts SOD1 dual expression vectors and uses thereof
US11434502B2 (en) 2017-10-16 2022-09-06 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (ALS)
US11931375B2 (en) 2017-10-16 2024-03-19 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (ALS)
WO2019094253A1 (en) 2017-11-08 2019-05-16 Avexis Inc. Means and method for preparing viral vectors and uses of same
WO2023073526A1 (en) 2021-10-25 2023-05-04 Novartis Ag Methods for improving adeno-associated virus (aav) delivery
WO2024015794A3 (en) * 2022-07-12 2024-03-07 The Research Institute At Nationwide Children's Hospital Adeno-associated virus gene therapy products and methods

Also Published As

Publication number Publication date
LT3702466T (lt) 2023-02-27
US20210108209A1 (en) 2021-04-15
PT3702466T (pt) 2023-01-24
WO2015031392A9 (en) 2015-06-18
RS63942B1 (sr) 2023-02-28
CA2922500A1 (en) 2015-03-05
SI3039146T1 (sl) 2020-10-30
HUE050546T2 (hu) 2020-12-28
LT3039146T (lt) 2020-07-27
ES2936220T3 (es) 2023-03-15
PT3039146T (pt) 2020-08-11
DK3039146T3 (da) 2020-08-10
EP4219727A3 (en) 2023-09-06
DK3702466T3 (da) 2023-03-20
CY1123272T1 (el) 2021-12-31
HRP20200862T1 (hr) 2020-08-21
EP3039146B1 (en) 2020-05-27
FI3702466T3 (fi) 2023-03-23
ES2799437T3 (es) 2020-12-17
SI3702466T1 (sl) 2023-04-28
EP3702466A1 (en) 2020-09-02
RS60434B1 (sr) 2020-07-31
HRP20230015T1 (hr) 2023-02-17
EP3702466B1 (en) 2022-12-28
EP4219727A2 (en) 2023-08-02
PL3702466T3 (pl) 2023-04-24
HUE061629T2 (hu) 2023-07-28
EP3039146A1 (en) 2016-07-06
WO2015031392A1 (en) 2015-03-05
US20190144868A1 (en) 2019-05-16
US10793861B2 (en) 2020-10-06

Similar Documents

Publication Publication Date Title
US10793861B2 (en) Products and methods for treatment of familial amyotrophic lateral sclerosis
ES2946747T3 (es) Variantes de cápsides de virus adenoasociado y su utilización para inhibir la angiogénesis
ES2941502T3 (es) Variantes de cápsides de virus adenoasociado y procedimientos de utilización de las mismas
AU2021261838A1 (en) Gene therapy for juvenile batten disease
US11773392B2 (en) AAV treatment of Huntington's disease
US11446395B2 (en) AAV vector for treatment of Friedreich's ataxia
EP2634253B1 (en) Adeno-associated virus virions for transferring genes into neural cells
JP7226801B2 (ja) 標的遺伝子導入のための方法および組成物
CN111718947B (zh) 用于治疗ⅲa或ⅲb型粘多糖贮积症的腺相关病毒载体及用途
US20190160187A1 (en) Gene therapy for the treatment of aldehyde dehydrogenase deficiency
CN114127296A (zh) Ube3a基因和表达盒及其应用
US20230058380A1 (en) Intrapancreatic m2 polarization of macrophages to treat type 1 diabetes

Legal Events

Date Code Title Description
AS Assignment

Owner name: RESEARCH INSTITUTE AT NATIONWIDE CHILDREN'S HOSPIT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KASPAR, BRIAN K.;FOUST, KEVIN;SIGNING DATES FROM 20140819 TO 20140820;REEL/FRAME:037850/0959

Owner name: LUDWIG INSTITUTE FOR CANCER RESEARCH, LTD, SWITZER

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CLEVELAND, DON W.;REEL/FRAME:037850/0975

Effective date: 20140822

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:CHILDREN'S HOSPITAL (COLUMBUS);REEL/FRAME:058788/0495

Effective date: 20220127