US20130039888A1 - Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders - Google Patents

Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders Download PDF

Info

Publication number
US20130039888A1
US20130039888A1 US13/491,326 US201213491326A US2013039888A1 US 20130039888 A1 US20130039888 A1 US 20130039888A1 US 201213491326 A US201213491326 A US 201213491326A US 2013039888 A1 US2013039888 A1 US 2013039888A1
Authority
US
United States
Prior art keywords
raav9
aav
polynucleotide
mice
genome
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
US13/491,326
Inventor
Douglas M. McCarty
Haiyan Fu
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.)
Nationwide Childrens Hospital Inc
Original Assignee
Nationwide Childrens Hospital 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 Nationwide Childrens Hospital Inc filed Critical Nationwide Childrens Hospital Inc
Priority to US13/491,326 priority Critical patent/US20130039888A1/en
Publication of US20130039888A1 publication Critical patent/US20130039888A1/en
Assigned to NATIONWIDE CHILDREN'S HOSPITAL, INC. reassignment NATIONWIDE CHILDREN'S HOSPITAL, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FU, HAIYAN, MCCARTY, DOUGLAS M.
Priority to US13/922,915 priority patent/US20130323207A1/en
Priority to US14/950,387 priority patent/US20160175406A1/en
Priority to US15/903,861 priority patent/US20180193431A1/en
Priority to US16/293,196 priority patent/US11590210B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • 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
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • C12N9/14Hydrolases (3)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14121Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14171Demonstrated in vivo effect
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y310/00Hydrolases acting on sulfur-nitrogen bonds (3.10)
    • C12Y310/01Hydrolases acting on sulfur-nitrogen bonds (3.10) acting on sulfur-nitrogen bonds (3.10.1)
    • C12Y310/01001N-Sulfoglucosamine sulfohydrolase (3.10.1.1)

Definitions

  • the present invention relates to methods and materials useful for systemically delivering polynucleotides across the blood brain barrier using adeno-associated virus as a vector.
  • the present invention relates to methods and materials useful for systemically delivering ⁇ -N-acetylglucosamidinase polynucleotides to the central and peripheral nervous systems, as well as the somatic system. Use of these methods and materials is indicated, for example, for treatment of the lysosomal storage disorder mucopolysaccharidosis IIIB.
  • the present invention relates to methods and materials useful for systemically delivering N-sulphoglucosamine sulfphohydrolase polynucleotides to the central and peripheral nervous systems, as well as the somatic system.
  • Use of this second type of methods and materials is indicated, for example, for treatment of the lysosomal storage disorder mucopolysaccharidosis IIIA.
  • Mucopolysaccharidosis (MPS) IIIB is a devastating lysosomal storage disease (LSD) caused by autosomal recessive defects in the gene coding a lysosomal enzyme, ⁇ -N-Acetylglucosaminidase (NAGLU).
  • LSD lysosomal storage disease
  • NAGLU ⁇ -N-Acetylglucosaminidase
  • GAG biologically important glycosaminoglycan
  • Cells throughout the CNS are particularly affected, resulting in complex secondary neuropathology.
  • MPS IIIB infants appear normal at birth, but develop progressive neurological manifestations that lead to premature death. Somatic manifestations of MPS IIIB occur in all patients, and involve virtually all organs, although they are mild relative to other forms of MPS, such as MPS I, II and VII.
  • MPS IIIA is a related LSD caused by autosomal recessive defects in the gene encoding a lysosomal enzyme, N-sulphoglucosamine sulphohydrolase (SGSH).
  • SGSH N-sulphoglucosamine sulphohydrolase
  • the lack of SGSH activity also disrupts the stepwise degradation of a class of biologically important GAG, leading to the accumulation of heparin sulfate oligosaccharides in lysosomes in cells of most tissues.
  • MPS IIIB No treatment is currently available for MPS IIIB or IIIA.
  • therapies have historically been limited to supportive care and management of complications.
  • MPS IIIB is not amenable to either hematopoietic stem cell transplantation or recombinant enzyme replacement therapy. These have instead been used to treat mostly somatic disorders in patients with MPS I, II and IV. This is because the neuropathology of MPS IIIB is global and the blood brain barrier (BBB) precludes effective central nervous system (CNS) access.
  • BBB blood brain barrier
  • the BBB is completely formed at birth in humans. In general, the BBB protects the CNS by selectively regulating the transport of molecules/agents from the blood circulation into the CNS or vice versa. Likewise, it prevents potential therapeutics from entering the CNS.
  • the BBB remains the most critical challenge to developing therapies for CNS diseases, especially global CNS disorders.
  • AAV vector system is one system with demonstrated therapeutic effect in a great variety of disease models. To date, no known pathogenesis has been linked to AAV in humans.
  • Recombinant AAV (rAAV) vectors based on AAV serotype 2 (AAV2) have been used in numerous studies for neurological diseases, transducing both neuronal and non-neuronal cells in the CNS with demonstrated therapeutic benefits in treating MPS and other LSDs in animals and in patients with Parkinson's and Batten's disease.
  • the BBB may still be permeable in neonatal mice while closed at birth in humans.
  • pretreatment with an N infusion of mannitol transiently disrupting the BBB facilitated the CNS entry of IV-delivered rAAV2, resulting in diffuse global CNS transduction and neurological correction.
  • McCarty et al. Gene Ther., 16: 1340-1352 (2009).
  • Adeno-associated virus 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, 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 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, 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 and AAV11.
  • AAV9 is described in U.S. Pat. No. 7,198,951 and in Gao et al., J. Virol., 78: 6381-6388 (2004).
  • 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.
  • the present invention provides methods and materials useful for systemically delivering polynucleotides such as NAGLU polynucleotides or SGSH polynucleotides across the BBB.
  • gene delivery is achieved by utilizing, for example, AAV serotype 9 (AAV9).
  • AAV9 AAV serotype 9
  • Vectors based on this serotype or functionally-related serotypes are able to cross the BBB unaided in neonate and adult animals.
  • An added benefit to using AAV9 vectors is that pre-existing immunity is less common than for AAV2 serotype.
  • the use of rh74 serotype AAV vectors among others is also contemplated by the invention.
  • the invention provides a method of delivering a NAGLU polynucleotide across the BBB comprising systemically administering a rAAV9 with a genome including the polynucleotide to a patient.
  • the rAAV9 genome is a single-stranded genome.
  • the present invention provides methods and materials useful for systemically delivering NAGLU polynucleotides across the blood brain barrier to the central and peripheral nervous system.
  • a method is provided of delivering a polynucleotide to the central nervous system comprising systemically administering a rAAV9 with a single-stranded genome including the genome to a patient.
  • a method of delivering a NAGLU polynucleotide to the peripheral nervous system comprising systemically administering a rAAV9 with a single-stranded genome including the polynucleotide to a patient is provided.
  • the NAGLU polynucleotide is delivered to brain. In some embodiments, the polynucleotide is delivered to the spinal cord. In some embodiments, the NAGLU polynucleotide is delivered to a lower motor neuron. In some embodiments, the polynucleotide is delivered to nerve and glial cells. In some embodiments, the glial cell is a microglial cell, an oligodendrocyte or an astrocyte. In some, embodiments, the rAAV9 is used to deliver a NAGLU polynucleotide to a Schwann cell.
  • NAGLU methods and materials are indicated, for example, for treating Sanfilippo syndrome Type B/MPS IIIB.
  • the invention provides a method of delivering an SGSH polynucleotide across the BBB comprising systemically administering a rAAV9 with a genome including the polynucleotide to a patient.
  • the rAAV9 genome is a self-complementary genome.
  • the rAAV9 genome is a single-stranded genome.
  • the present invention provides methods and materials useful for systemically delivering SGSH polynucleotides across the blood brain barrier to the central and peripheral nervous system.
  • a method is provided of delivering a polynucleotide to the central nervous system comprising systemically administering a rAAV9 with a self-complementary genome including the genome to a patient.
  • a method of delivering a SGSH polynucleotide to the peripheral nervous system comprising systemically administering a rAAV9 with a self-complementary genome including the polynucleotide to a patient is provided.
  • the SGSH polynucleotide is delivered to brain. In some embodiments, the polynucleotide is delivered to the spinal cord. In some embodiments, the SGSH polynucleotide is delivered to a lower motor neuron. In some embodiments, the polynucleotides is delivered to nerve and glial cells. In some embodiments, the glial cell is a microglial cell, an oligodendrocyte or an astrocyte. In some, embodiments, the rAAV9 is used to deliver a SGSH polynucleotide to a Schwann cell.
  • administration of the rAAV9 encoding a NAGLU or SGSH polypeptide is preceded by administration of mannitol.
  • the invention provides rAAV genomes comprising one or more AAV ITRs flanking a polynucleotide encoding a NAGLU.
  • the NAGLU polynucleotide is operatively linked to transcriptional control DNAs, specifically promoter DNA and polyadenylation signal sequence DNA that are functional in target cells to form a gene cassette.
  • the gene cassette may also include intron sequences to facilitate processing of an RNA transcript when expressed in mammalian cells.
  • the invention provides rAAV genomes comprising one or more AAV ITRs flanking a polynucleotide encoding an SGSH.
  • the SGSH polynucleotide is operatively linked to transcriptional control DNAs, specifically promoter DNA and polyadenylation signal sequence DNA that are functional in target cells to form a gene cassette.
  • the gene cassette may also include intron sequences to facilitate processing of an RNA transcript when expressed in mammalian cells.
  • 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-3 is provided in GenBank Accession No. NC — 1829
  • the complete genome of AAV-4 is provided in GenBank Accession No. NC — 001829
  • the AAV-5 genome is provided in GenBank Accession No. AF085716
  • the complete genome of AAV-6 is provided in GenBank Accession No. NC — 00 1862
  • at least portions of AAV-7 and AAV-8 genomes are provided in GenBank Accession Nos. AX753246 and AX753249, respectively
  • the AAV-9 genome is provided in Gao et al., J. Virol., 78: 6381-6388 (2004)
  • the AAV-10 genome is provided in Mol. Ther., 13(1): 67-76 (2006)
  • the AAV-11 genome is provided in Virology, 330(2): 375-383 (2004).
  • NAGLU polypeptides contemplated include, but are not limited to, a NAGLU polypeptide with the amino acid sequence set out in SEQ ID NO: 2.
  • SGSH polypeptides contemplated include, but are not limited to, a SGSH polypeptide with the amino acid sequence set out in SEQ ID NO: 4.
  • polypeptides contemplated include full-length proteins, precursors of full length proteins, biologically active subunits or fragments of full length proteins, as well as biologically active analogs (e.g., derivatives and variants) of any of these forms of polypeptides.
  • polypeptides include, for example, those that (1) have an amino acid sequence that has greater than about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% or about 99% or greater amino acid sequence identity, over a region of at least about 25, about 50, about 100, about 200, about 300, about 400, or more amino acids, to a polypeptide encoded by a nucleic acid or an amino acid sequence described herein.
  • biologically active derivative As used herein “biologically active derivative,” “biologically active fragment,” “biologically active analog” or “biologically active variant” includes any derivative or fragment or analog or variant of a molecule having substantially the same functional and/or biological properties of said molecule, such as enzymatic activities.
  • an “analog,” such as a “variant” or a “derivative,” is a compound substantially similar in structure to and having the same biological activity as, albeit in certain instances to a differing degree, a naturally-occurring molecule.
  • a “derivative,” for example, is a type of analog and refers to a polypeptide sharing the same or substantially similar structure as a reference polypeptide that has been modified, e.g., chemically.
  • a polypeptide variant for example, is a type of analog and refers to a polypeptide sharing substantially similar structure and having the same biological activity as a reference polypeptide (i.e., “native polypeptide” or “native therapeutic protein”).
  • Variants differ in the composition of their amino acid sequences compared to the naturally-occurring polypeptide from which the variant is derived, based on one or more mutations involving (i) deletion of one or more amino acid residues at one or more termini of the polypeptide and/or one or more internal regions of the naturally-occurring polypeptide sequence (e.g., fragments), (ii) insertion or addition of one or more amino acids at one or more termini (typically an “addition” or “fusion”) of the polypeptide and/or one or more internal regions (typically an “insertion”) of the naturally-occurring polypeptide sequence or (iii) substitution of one or more amino acids for other amino acids in the naturally-occurring polypeptide sequence.
  • Variant polypeptides include insertion variants, wherein one or more amino acid residues are added to a therapeutic protein amino acid sequence of the present disclosure. Insertions may be located at either or both termini of the protein, and/or may be positioned within internal regions of the therapeutic protein amino acid sequence. Insertion variants, with additional residues at either or both termini, include for example, fusion proteins and proteins including amino acid tags or other amino acid labels.
  • one or more amino acid residues in a therapeutic protein polypeptide as described herein are removed.
  • Deletions can be effected at one or both termini of the therapeutic protein polypeptide, and/or with removal of one or more residues within the therapeutic protein amino acid sequence.
  • Deletion variants therefore, include fragments of a polypeptide sequence.
  • substitution variants one or more amino acid residues of a therapeutic protein polypeptide are removed and replaced with alternative residues.
  • the substitutions are conservative in nature and conservative substitutions of this type are well known in the art.
  • the present disclosure embraces substitutions that are also non-conservative. Exemplary conservative substitutions are described in Lehninger, [Biochemistry, 2nd Edition; Worth Publishers, Inc., New York (1975), pp. 71-77] and are set out immediately below.
  • 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 Production of rAAV requires that the following components are present within a single cell (denoted herein as a packaging cell): a rAAV genome, AAV rep and cap genes separate from (i.e., not in) the rAAV genome, and 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.
  • Production of pseudotyped rAAV is disclosed in, for example, WO 01/83692 which is incorporated by reference herein in its 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.
  • rAAV infectious encapsidated rAAV particles
  • the rAAV genome is a self-complementary genome.
  • the invention includes, but is not limited to, the exemplified rAAV named “rAAV9-CMV-hNAGLU.”
  • the rAAV genome has in sequence an AAV2 ITR, the cytomegalovirus (CMV) immediate early promoter/enhancer, an SV40 intron (SD/SA), the NAGLU DNA set out in SEQ ID NO: 1, a polyadenylation signal sequence from bovine growth hormone and another AAV2 ITR.
  • the DNA sequence of the vector genome is set out in SEQ ID NO: 5.
  • the genome lacks AAV rep and cap DNA, that is, there is no AAV rep or cap DNA between the ITRs of the genome.
  • the invention also includes, but is not limited to, rAAV encoding SGSH.
  • the rAAV genome has in sequence an AAV2 ITR, the CMV immediate early promoter/enhancer, the SGSH DNA set out in SEQ ID NO: 3, a polyadenylation signal sequence from bovine growth hormone and a AAV2 ITR lacking the terminal resolution site.
  • the rAAV genome has in sequence an AAV2 ITR, the mouse U1a promoter, the SGSH DNA set out in SEQ ID NO: 3, a polyadenylation signal sequence from bovine growth hormone and a AAV2 ITR lacking the terminal resolution site.
  • rAAV genome has in sequence an AAV2 ITR, the mouse U1a promoter, an intron, the SGSH DNA set out in SEQ ID NO: 3, a polyadenylation signal sequence from bovine growth hormone and a AAV2 ITR lacking the terminal resolution site.
  • the genomes lack AAV rep and cap DNA, that is, there is no AAV rep or cap DNA between the ITRs of the genomes.
  • NAGLU and SGSH DNAs include, without limitation, those that (1) hybridize under stringent hybridization conditions to a nucleic acid encoding an amino acid sequence as described herein, and conservatively modified variants thereof; (2) have a nucleic acid sequence that has greater than about 95%, about 96%, about 97%, about 98%, about 99%, or higher nucleotide sequence identity, over a region of at least about 25, about 50, about 100, about 150, about 200, about 250, about 500, about 1000, or more nucleotides (up to the full length sequence of the mature protein), to a nucleic acid sequence as described herein.
  • Exemplary “stringent hybridization” conditions include hybridization at 42° C.
  • the rAAV 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 encoding an NAGLU polypeptide. These compositions may be used to treat mucopolysaccharidosis IIIB. In other embodiments, compositions of the present invention may include two or more rAAV encoding different polypeptides of interest.
  • compositions comprising rAAV of the present invention encoding an SGSH polypeptide. These compositions may be used to treat mucopolysaccharidosis IIIA. In other embodiments, compositions of the present invention may include two or more rAAV encoding different polypeptides of interest.
  • 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 counterions such as sodium; and/or nonionic surfactants such as T
  • 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 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. 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.
  • DNase resistant particles DNase resistant particles
  • These dosages of rAAV may range from about 1 ⁇ 10 11 vg/kg, 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.
  • the dosages of rAAV may range from about 1 ⁇ 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.
  • Treatment by methods of the invention comprises the step of administering an intravenous (IV) effective dose, or effective multiple doses, of a composition comprising a rAAV of the invention to an animal (including a human being) in need thereof. If the dose is administered prior to development of a disorder/disease, the administration is prophylactic. If the dose is administered after the development of a disorder/disease, the administration is therapeutic.
  • IV intravenous
  • 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.
  • 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., transient or long-term immunosuppression
  • compositions suitable for systemic (IV) use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of a dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal and the like. In many cases it will be preferable to include isotonic agents, for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by use of agents delaying absorption, for example, aluminum monostearate and gelatin, and Tween family of products (e.g., Tween 20).
  • Sterile injectable solutions are prepared by incorporating rAAV in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filter sterilization.
  • dispersions are prepared by incorporating the sterilized active ingredient into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and the freeze drying technique that yield a powder of the active ingredient plus any additional desired ingredient from the previously sterile-filtered solution thereof.
  • Transduction of cells with rAAV of the invention results in sustained expression of NAGLU or SGSH polypeptide.
  • Transduction may be carried out with gene cassettes comprising tissue specific control elements, for example, promoters that allow expression specifically within neurons or specifically within astrocytes. Examples include neuron specific enolase and glial fibrillary acidic protein promoters. Inducible promoters under the control of an ingested drug may also be developed.
  • a polynucleotide delivered using the materials and methods of the invention can be placed under regulatory control using systems known in the art.
  • systems such as the tetracycline (TET on/off) system [see, for example, Urlinger et al., Proc. Natl. Acad. Sci. USA 97(14):7963-7968 (2000) for recent improvements to the TET system] and Ecdysone receptor regulatable system [Palli et al., Eur J. Biochem 270: 1308-1315 (2003] may be utilized to provide inducible polynucleotide expression.
  • TET on/off tetracycline
  • Ecdysone receptor regulatable system Ecdysone receptor regulatable system
  • the invention provides methods of systemically administering an effective dose (or doses, administered essentially simultaneously or doses given at intervals) of rAAV of the invention to a patient in need thereof.
  • FIG. 1 is a map of the rAAV-CMV-hNAGLU vector genome.
  • FIG. 2 shows improved behavior and extended survival in MPS IIIB mice after systemic gene transfer by rAAV-CMV-hNAGLU.
  • FIG. 2 c Survival (i 5/group, P ⁇ 0.001). +/+: wt; ⁇ / ⁇ : MPS IIIB; AAV9-L, AAV9-H: MPS IIIB mice treated with 5 ⁇ 10 12 or 1.5 ⁇ 10 13 vg/kg rAAV9-hNAGLU vector, respectively.
  • AAV9-L ⁇ : P ⁇ 0.05 (vs. AAV9-H); &: P>0.05 (vs. ⁇ / ⁇ ).
  • FIG. 3 a Dose-response. +/+: wt; AAV9-11, AAV9-L: MPS IIIB mice treated with 1.5 ⁇ 10 13 (AAV9-H) or 5 ⁇ 10 12 vg/kg (AAV9-L) vector;
  • FIG. 3 b Impact of mannitol pretreatment.
  • FIG. 4 shows the significant reduction of GAG content in the CNS and somatic tissues. Tissues from MPS IIIB mice treated with rAAV9-hNAGLU were assayed to quantify GAG content (6 and 9 mo pi).
  • FIG. 4 a Dose response.
  • FIG. 4 b Impact of mannitol pretreatment. +/Ai wt; ⁇ / ⁇ : MPS IIIB; AAV9-H, AAV9-L: MPS MB mice treated with 1.5 ⁇ 1013 vg or 5 ⁇ 1012 vg/kg vector; M+, M ⁇ : MPS IIIB mice treated with rAAV9 vector (2 ⁇ 1013 vg/kg) with or without mannitol pretreatment.
  • FIG. 5 shows rAAV9-mediated correction of astrocytosis and neurodegeneration in MPS IIIB mice.
  • Brain sections of MPS IIIB mice treated with rAAV9-CMV-hNAGLU vector (6 mo pi) were assayed for GFAP by immunofluorescence and stained with toluidine blue for histopathology.
  • FIG. 5 a Number of astrocytes: Data are means ⁇ SD of GFAP+ cells per 330 ⁇ 433 pm on 6-8 IF-GFAP-staining sections/mouse, from 3 mice/group.
  • FIG. 5 b shows rAAV9-mediated correction of astrocytosis and neurodegeneration in MPS IIIB mice.
  • Number of purkinje cells Data are means ⁇ SD of purkinje cells/200 p.m (in length) in ansiform lobules in cerebellum on 6 toluidine blue stained sections/mouse, from 3 mice/group.
  • NT non-treated MPS IIIB mouse
  • AAV9 MPS IIIB mouse treated with rAAV9.
  • CTX cerebral cortex
  • ST Striatum
  • TH thalamus
  • BS Brain stem. *: P ⁇ 0.01 vs. non-treated.
  • FIG. 6 shows rAAV9-mediated expression of functional rSGSH in tissues of treated MPSIIIA mice. For each tissue, AAV9, rh74 and untreated result bars are respectively shown from left to right.
  • FIG. 7 shows a significant reduction of GAG content in tissues of treated MPSIIIA mice. For each tissue, AAV9, rh74 and untreated result bars are respectively shown from left to right.
  • FIG. 8 shows an improvement in cognitive behavior assays after treatment of one-month old MPSIIIA mice with low dose scAAV9 or rh74-U1a-SGSH.
  • scAAV9 or rh74-U1a-SGSH low dose scAAV9 or rh74-U1a-SGSH.
  • untreated, wild type and either AAV9 or rh74 result bars are respectively shown from left to right.
  • FIG. 9 shows a significant reduction of GAG content in tissues of MPSIIIA mice treated at 2 or 6 months of age. For each tissue, wild type, untreated, two-month and six-month result bars are respectively shown from left to right.
  • FIG. 10 shows an improvement in cognitive behavior assays after treatment of two- or six-month old MPSIIIA mice with high dose scAAV9 or rh74-U1a-SGSH.
  • scAAV9 or rh74-U1a-SGSH high dose scAAV9 or rh74-U1a-SGSH.
  • untreated, wild type and either AAV9 or rh74 result bars are respectively shown from left to right.
  • Example 1 describes rAAV encoding hNAGLU.
  • Example 2 describes the administration of the rAAV encoding hNAGLU to MPSIIIB mice.
  • Examples 3 through 6 describe the beneficial results of administration of the rAAV.
  • Example 7 discusses the significance of the results.
  • Example 8 describes rAAV encoding SGSH.
  • Examples 9 through 11 describe administration of various dosages of rAAV encoding SGSH to MPSIIIA mice of varying ages, as well as the beneficial effects of the administration.
  • a rAAV vector plasmid containing AAV2 ITRs, an immediate early CMV promoter/enhancer, an SV40 intron, a human ⁇ -N-acetylglucosaminidase coding region, a bGH polyadenylation signal sequence, and ampicillin resistance gene, was used to produce a rAAV9-CMV-hNAGLU viral vector.
  • FIG. 1 is a map of the vector genome wherein the length of the various elements of the genome is indicated below the element.
  • a control self-complementary AAV encoding green fluorescent protein, scAAV9-CMV-GFP was also produced, containing AAV2 terminal repeats, a human cytomegalovirus (CMV) immediate-early promoter, SV40 splice donor/acceptor signal, a eGFP coding sequence, and SV40 polyadenylation signal.
  • CMV human cytomegalovirus
  • CMV-GFP cytomegalovirus-green fluorescent protein
  • the rAAV9-CMV-hNaGlu-treated MPS BIB mice and controls were tested for behavioral performance at approximately 5.0-5.5 months of age as follows.
  • mice treated IV with 5 ⁇ 10 12 or 1.5 ⁇ 10 13 vg/kg rAAV9-NAGLU were tested for behavior at 5-5.5 mo of age to assess the neurological impacts. Both dosage groups exhibited significant decreases in latency to find a hidden platform in a water maze ( FIG. 2 a ), and significantly longer latency to fall from an accelerating rotarod ( FIG. 2 b ), compared with non-treated MPS IIIB mice, indicating the correction of cognitive and motor function. There were no significant differences in behavior performance between these two dose groups.
  • mice were continuously observed for the development of endpoint symptoms, or until death occurred.
  • the endpoint was when the symptoms of late stage clinical manifestation (urine retention, rectal prolapse, protruding penis) in MPS IIIB mice became irreversible, or when wt control mice were 24 months or older.
  • Longevity data were analyzed using Kaplan-Meier method. The significance level was set at P ⁇ 0.05. Results are shown in FIG. 2 c.
  • tissue analyses were carried out at 6 mo and 9 mo post injection (pi). Mice were anesthetized with 2.5% Avertin before tissue collection. Brain, spinal cord and multiple somatic tissues were collected on dry ice or embedded in OCT compound and stored at ⁇ 70° C., before being processed for analyses. Tissues were also processed for paraffm sectioning.
  • Tissue samples from scAAV9-GFP vector-treated mice were collected for analysis 4-5 weeks pi.
  • the mice were anesthetized with 2.5% Avertin and then perfused transcardially with cold PBS (0.1M, pH7.4), followed by 4% paraformaldehyde in phosphate buffer (0.1M, pH7.4).
  • the entire brain and spinal cord, as well as multiple somatic tissues were collected and fixed in 4% paraformaldehyde overnight at 4° C. before being further processed for vibratome sectioning.
  • Tissues were analyzed at 6 mo and/or 9 mo pi by NAGLU activity assay to determine the distribution and level of rAAV9-mediated transgene expression.
  • Tissue samples were assayed for NaGlu enzyme activity following a published procedure with modification.
  • the assay measures 4-methylumbelliferone (4MU), a fluorescent product formed by hydrolysis of the substrate 4-methylumbellireyl-N-acetyla-D-glucosaminide.
  • the NaGlu activity is expressed as unit/mg protein. 1 unit is equal to 1 nmol 4MU released/h at 37° C. Results are shown in FIG. 3 .
  • GAG was extracted from tissues following published procedures [van de Lest et al., Anal. Biochem. 221: 356-361(1994)] with modification [Fu et al., Gene Ther., 14: 1065-1077 (2007).
  • Dimethylmethylene blue (DMB) assay was used to measure GAG content [de Jong et al., Clin. Chem., 35: 1472-1477 (1989)].
  • the GAG samples (from 0.5-1.0 mg tissue) were mixed with H 2 O to 40 ml before adding 35 nM DMB (Polysciences CEO 03610-1) in 0.2 mM sodium formate buffer (SFB, pH 3.5). The product was measured using a spectrophotometer (0D535).
  • the GAG content was expressed as ⁇ g/mg tissue.
  • Urine GAG content was also measured. Heparan sulfate (Sigma, H9637) was used as standard. Results are shown in FIG. 4 .
  • Immunofluorescence was performed to identify cells expressing hNAGLU, GFP or glial fibrillary acidic protein (GFAP) for astrocytes, using antibodies against hNaGlu (a kind gift from Dr. EF Neufeld, UCLA), GFP (Invitrogen) or GFAP (Chemicon), and corresponding secondary antibody conjugated with AlexaFluor 568 or AlexaFluor 488 (Molecular Probes).
  • the IF staining was performed on thin cryostat sections (8 p.m) of tissue samples following procedures recommended by the manufacturers. The sections were visualized under a fluorescence microscope.
  • Tissues were assayed for histopathology to visualize the impact of IV rAAV9-NAGLU gene delivery on the lysosomal storage pathology in MPS IIIB mice. Histopathology was performed following standard methods. Paraffm sections (41.un) were fixed with 4% paraformaldehyde in phosphate buffer (0.1 M, pH 7.2) at 4° C. for 15 min and stained with 1% toluidine blue at 37° C. for 30 min to visualize lysosomal GAG. The sections were mounted, and imaged under a light microscope.
  • Taqman primers specific for the CMV promoter were used to detect rAAV vector genomes: f: GGCAGTACATC AAGTGTATC (SEQ ID NO: 6); r: ACCAATGG TAATAG CGATGAC (SEQ ID NO: 7); probe: [6 ⁇ FAM]AATGACGGTAAAT GGCCCGC[TAMRA ⁇ 6 ⁇ FAM] (SEQ ID NO: 8).
  • Genomic DNA was quantified in parallel samples using ⁇ -actin specific primers: f: GTCATCAC TATTG GCAACGA (SEQ ID NO: 9); r: CTCAGGAGTTTTGTCACCTT (SEQ ID NO: 10); probe: [6 ⁇ FAM]TTCCGATGCCCT GAGGCTCT[Tamra ⁇ Q] (SEQ ID NO: 11).
  • Genomic DNA from nontreated MPS IIIB mouse tissues was used as controls or background and absence of contamination. Global CNS and widespread somatic restoration of NAGLU.
  • Tissues were analyzed at 6 months and/or 9 months pi by immunofluorescence (IF) and NAGLU activity assay to determine the distribution and level of rAAV9-mediated transgene expression.
  • IF immunofluorescence
  • NAGLU-specific IF was detected throughout the brains of treated mice, in neurons, glia, and abundant endothelial cells in capillaries and larger blood vessels, in an apparently dose—dependent fashion. No significant differences were observed in the distribution or levels of rNaGlu signal between 6 months and 9 months pi.
  • NAGLU-positive glial cells were not costained with anti-glial fibrillary acidic protein (GFAP) Ab, and were likely to be oligodendrocytes, based on their morphology.
  • GFAP anti-glial fibrillary acidic protein
  • rNAGLU protein was detected in 20-40% of hepatocytes, >95% of cardiomyocytes, and 10-30% of skeletal myocytes.
  • the distribution of rAAV9-transduced hepatocytes was uniform throughout the liver. Transduction in abundant neurons in myenteric plexus and submucosal plexus of the intestine was observed, suggesting efficient targeting of the peripheral nervous system (PNS).
  • PNS peripheral nervous system
  • the rNAGLU signals were mostly present in granules, whereas scAAV9-mediated GFP signals were uniform in the cytoplasm of transduced cells, suggesting correct lysosomal trafficking of rNAGLU. Transduction of endothelial cells was also observed in peripheral tissues of rAAV9-GFP vector-treated mice.
  • Transgene enzymatic activity was assayed to quantify the expression and the functionality of rAAV9-mediated rNAGLU. There were no significant differences in tissue NAGLU activity at 6 and 9 months pi, suggesting stable transduction.
  • the rAAV mediated rNaGlu was metabolically functional and the tissue rNAGLU activity was dose-dependent, with approximately normal levels in the brains of mice receiving 5 ⁇ 10 12 vg/kg vector, and supra-physiologic levels in the brains of mice receiving 1.5 ⁇ 10 13 vg/kg ( FIG. 3 a ).
  • NAGLU activity at normal or subnormal levels in the liver, lung and intestine ( FIG. 3 a ), supra-physiologic levels in the skeletal muscles ( FIG.
  • Tissues were assayed for GAG content to quantify the impact of IV rAAV9-NAGLU gene delivery on the lysosomal storage pathology in MPS IIIB mice.
  • the single IV rAAV9-NAGLU injection led to a reduction of GAG content to normal levels in the brain, liver, heart, lung, intestine and skeletal muscle in mice of all four treatment groups ( FIG. 4 ).
  • Doses of 5 ⁇ 10 12 ⁇ g or 1.5 ⁇ 10 13 vg/kg resulted in partial GAG reduction in the spleen but had no impact in kidney ( FIG. 4 a ).
  • Histopathology showed complete clearance or reduction of lysosomal storage lesions in the vast majority of CNS areas, including cerebral cortex, thalamus, brain stem, hippocampus, and spinal cord in all four treatment groups. There were decreases in the size, number of vacuoles, and number of cells with lysosomal storage lesions, even in the few brain areas that did not show a complete correction, such as purkinje cells and cells in the striatum and hypothalamus. Importantly, the majority of brain and spinal cord parenchymal cells exhibited a well defined normalized morphology.
  • Quantitative real-time PCR was performed to compare the amount of rAAV9-CMV-hNaglu vector entering the CNS versus somatic tissues.
  • Table 1 shows the distribution of the vector genome in different tissues/organs of MPS IIIB mice treated with IV vector injection at varying doses. The highest concentrations of vector genome were detected in liver (8.20 ⁇ 4.73-32.09 ⁇ 3.93 copies/cell), followed by heart (0.07-0.22 copies/cell), and brain (0.06 ⁇ 0.001-0.15 ⁇ 0.02 copies/cell), and very low copy numbers were detected in other tissues/organs (Table 1).
  • This differential vector distribution in rAAV9-treated MPS IIIB mice largely correlated with the distribution of rNAGLU IF and enzymatic activity.
  • mannitol pretreatment increased the vector copy numbers in the brain, correlating with brain NAGLU activity levels. Furthermore, these data reflect persistence of vector genome distribution in treated mice at 6 months pi, supporting a stable long-term transduction. Levels of vector genome copies correlating with rNAGLU activity and distribution were not detectable, possibly due to difficulties in quantitative isolation of DNA from muscle tissue.
  • rAAV9-L IV infusion of 5 ⁇ 10 12 vg/kg
  • rAAV9-H IV infusion of 1.5 ⁇ 10 13 vg/kg
  • rAAV9-M ⁇ IV infusion of 2 ⁇ 10 13 vg/kg without mannitol pretreatment
  • rAAV9-M+ IV infusion of 2 ⁇ 10 13 vg/kg following mannitol pretreatment. *Data from 1 sample in duplicates.
  • the clinically meaningful therapeutic benefits of the IV-delivered rAAV9 vector in MPS IIIB mice were achieved at a lower dose than the mannitol-facilitated, systemically delivered rAAV2 vector.
  • the enhanced rAAV9-CNS transduction in response to mannitol pretreatment suggests further potential for reducing the vector dose, and the attendant risk and burden to patients.
  • the IV vector injection resulted in a ubiquitously diffuse, global rAAV9-NaGlu transduction throughout the CNS, reflecting the expected distribution pattern for vascular delivery. This contrasts sharply with the focal gradient distribution typically achieved through direct brain parenchymal injection, or the periventricular transduction pattern from intracisternal and intraventricular injection. While similar to the pattern of transgene expression from IV-delivered rAAV2 after mannitol pretreatment, the extent of rAAV9 transduction was significantly higher in all areas of the brain. This correlates with the increased effects on longevity and cognitive function compared to that previously achieved using rAAV2-mannitol treatment, and the normal or above normal levels of NAGLU activity in the CNS. These findings strongly support the use of the trans-BBB neurotropic rAAV9 as a vector for CNS gene therapy and reinforce the view that efficient CNS delivery is the most critical issue for developing therapies to treat MPS IIIB.
  • the rAAV9-transduced CNS cells include neurons, glia and endothelia. Neuronal cell transduction appears to be non-preferential, including most types of neurons throughout the brain. In contrast, the transduction of glial cells appears to be cell-type specific, targeting predominantly oligodendrocyte-like cells, though it is unclear whether this is a receptor- or promoter-specific phenomenon. In a previous report [Faust et al., supra] describing predominant transduction of astocytes after systemic injection of rAAV9 vector in adult mice, a hybrid chicken J3-actin/CMV-enhancer promoter was used, rather than the CMV enhancer-promoter used in the present study.
  • lysosomal protein In normal cells, 5-20% of newly synthesized lysosomal protein is secreted and available to be taken up by neighboring cells, leading to the by-stander effect.
  • the abundant transduction of endothelial cells in the brain may be an important contributor to the effectiveness of rAAV9 gene delivery for MPS IIIB because of the close association between CNS cells and brain microvascular endothelial cells, which together constitute the neurovascular unit. While the observed high levels of rNAGLU expression stem from the transduction of a relatively small number of CNS cells, it is sufficient to correct the neuropathology leading to functional correction of the neurological disorders.
  • the rAAV9 treatment also led to a regular morphology in CNS cells, and the correction of major secondary neuropathology, astrocytosis, and neurodegeneration. It is worth noting that this level of correction of CNS pathology was not achieved in previous studies using rAAV2-hNAGLU vector with mannitol. While neuropathology is the primary cause of mortality in MPS IIIB patients, somatic correction may provide additional therapeutic benefits, since lysosomal storage pathology inevitably manifests in virtually all organs.
  • the IV-delivered rAAV9 exhibited broad tropism in peripheral tissues in a distinct pattern, as previously reported, reflecting extensive extravasation and cell-type specific transduction.
  • the primary source of circulating NAGLU may be liver, muscle, or endothelium.
  • the decrease in plasma levels in response to mannitol pretreatment correlated with decreased transduction in muscle rather than liver, suggesting that liver may not be the primary source.
  • neurotropism is a general property of the AAV9 serotype, and not dependent on the specific structure of the brain neurovascular unit. Broad neurotropism is a valuable property in gene therapy for the treatment of MPS IIIB, considering that lysosomal storage pathology manifests not only in the CNS but also in the PNS.
  • a rAAV vector plasmid was used to produce three different rAAV9-CMV-hSGSH viral vectors.
  • the three self-complementary AAV hSGSH vector-producing plasmids were constructed using conventional plasmid cloning techniques.
  • Each vector genome contains an SGSH coding region (SEQ ID NO: 3) and either the mouse U1a promoter, with or without an intron, or a CMV promoter without an intron,
  • Each vector genome also contains a bGH polyadenylation signal.
  • Each self-complementary vector plasmid construct contains one intact AA2 terminal repeat and one modified AAV2 terminal repeat missing the terminal resolution site, thereby forcing the replication of dimeric self-complementary DNA genomes.
  • Self-complementary AAV hSGSH viral vectors were produced and packaged in AAV serotype 9 capsids. The viral vectors were tested for expression of hSGSH protein and reduction of GAG storage in human MPS IIIA fibroblasts.
  • MPS IIIA mice were injected at 10 weeks of age with 5 ⁇ 10 12 vgp/kg) of scAAV-U1a-hSGSH vector encapsidated in either AAV9 or AAVrh74 serotype. At 10 days post-injection, the mice were euthanized and assays were performed to determine the effects of the treatment.
  • FIG. 6 shows enzyme expression relative to untreated MPS IIIA mice at the same age ( ⁇ / ⁇ ).
  • the scAAV-SGSH vectors reached the CNS and expressed the transgene within days of administration.
  • FIG. 7 shows GAG content measured in the kidney (Kid), heart (Hrt), muscle (Mus), lung, brain, Liver (Liv) and spleen (Spl).
  • Sections of CNS and somatic tissues were stained with the lysosomal marker, Lamp1, revealing clearance of lysosomal storage pathology. Histopathology additionally revealed numerous clear vacuoles present in untreated mice but corrected in treated animals.
  • MPS IIIA Vector was administered by tail vein injection in MPS IIIA mice at one month of age at an approximately 25-fold lower dose than in Example 9.
  • MPS IIIA mice were treated with 1.7 ⁇ 10 11 vgp/kg scAAV9-U1a-hSGSH or 2.7 ⁇ 10 11 vgp/kg scAAVrh74-U1a-hSGSH vector.
  • MPS IIIA mice were treated with a high dose (5 ⁇ 10 12 vgp/kg) of scAAV9-U1a-hSGSH vector at 6 months of age, after significant neuropathology had already developed. At 7-7.5 months age, the animals were tested for learning ability in the Morris water maze. At 7.5 months of age, the mice were euthanized and tissues assayed for glycosaminoglycan (GAG) content. Tissues analyzed include liver (Liv), kidney (Kid), heart (Hrt), brain, spleen (Spl), lung, skeletal muscle, and intestine.
  • GAG glycosaminoglycan
  • FIG. 9 shows clearance of accumulated GAGs in different tissues, including CNS.
  • FIG. 10 shows, compared to untreated controls, treated animals were similar to wt mice in their latency to locate the hidden platform (upper charts) and spent more time in the zone (4) where the platform had been in the previous tests when the platform was removed (lower charts).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Mycology (AREA)
  • Obesity (AREA)
  • Diabetes (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present invention relates to methods and materials useful for systemically delivering polynucleotides across the blood brain barrier using adeno-associated virus as a vector. For example, the present invention relates to methods and materials useful for systemically delivering α-N-acetylglucosamidinase polynucleotides to the central and peripheral nervous systems, as well as the somatic system. Use of these methods and materials is indicated, for example, for treatment of the lysosomal storage disorder mucopolysaccharidosis IIIB. As another example, the present invention relates to methods and materials useful for systemically delivering N-sulphoglucosamine sulfphohydrolase polynucleotides to the central and peripheral nervous systems, as well as the somatic system. Use of this second type of methods and materials is indicated, for example, for treatment of the lysosomal storage disorder mucopolysaccharidosis IIIA.

Description

  • This application claims the benefit of the filing date of U.S. Provisional Patent Application No. 61/494,635 filed Jun. 8, 2011, which is incorporated by reference herein in its entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to methods and materials useful for systemically delivering polynucleotides across the blood brain barrier using adeno-associated virus as a vector. For example, the present invention relates to methods and materials useful for systemically delivering α-N-acetylglucosamidinase polynucleotides to the central and peripheral nervous systems, as well as the somatic system. Use of these methods and materials is indicated, for example, for treatment of the lysosomal storage disorder mucopolysaccharidosis IIIB. As another example, the present invention relates to methods and materials useful for systemically delivering N-sulphoglucosamine sulfphohydrolase polynucleotides to the central and peripheral nervous systems, as well as the somatic system. Use of this second type of methods and materials is indicated, for example, for treatment of the lysosomal storage disorder mucopolysaccharidosis IIIA.
  • INCORPORATION BY REFERENCE OF THE SEQUENCE LISTING
  • This application contains, as a separate part of disclosure, a Sequence Listing in computer-readable form (46031A_SeqListing.txt; 22,737 byte ASCII text file, created Jun. 7, 2012) which is incorporated by reference herein in its entirety.
  • BACKGROUND
  • Mucopolysaccharidosis (MPS) IIIB is a devastating lysosomal storage disease (LSD) caused by autosomal recessive defects in the gene coding a lysosomal enzyme, α-N-Acetylglucosaminidase (NAGLU). The lack of NAGLU activity disrupts the stepwise degradation of a class of biologically important glycosaminoglycan (GAG), leading to the accumulation of heparan sulfate oligosaccharides in lysosomes in cells of most tissues. Cells throughout the CNS are particularly affected, resulting in complex secondary neuropathology. MPS IIIB infants appear normal at birth, but develop progressive neurological manifestations that lead to premature death. Somatic manifestations of MPS IIIB occur in all patients, and involve virtually all organs, although they are mild relative to other forms of MPS, such as MPS I, II and VII.
  • MPS IIIA is a related LSD caused by autosomal recessive defects in the gene encoding a lysosomal enzyme, N-sulphoglucosamine sulphohydrolase (SGSH). The lack of SGSH activity also disrupts the stepwise degradation of a class of biologically important GAG, leading to the accumulation of heparin sulfate oligosaccharides in lysosomes in cells of most tissues.
  • No treatment is currently available for MPS IIIB or IIIA. For all of the MPS disorders, therapies have historically been limited to supportive care and management of complications. MPS IIIB is not amenable to either hematopoietic stem cell transplantation or recombinant enzyme replacement therapy. These have instead been used to treat mostly somatic disorders in patients with MPS I, II and IV. This is because the neuropathology of MPS IIIB is global and the blood brain barrier (BBB) precludes effective central nervous system (CNS) access.
  • For the majority of CNS diseases, effective treatments are rare since the CNS is located in a well protected environment and isolated by a highly defined anatomical/functional barrier. The BBB is completely formed at birth in humans. In general, the BBB protects the CNS by selectively regulating the transport of molecules/agents from the blood circulation into the CNS or vice versa. Likewise, it prevents potential therapeutics from entering the CNS. The BBB remains the most critical challenge to developing therapies for CNS diseases, especially global CNS disorders.
  • It is contemplated herein that gene therapy has potential for treating LSDs because the secretion of lysosomal enzymes, including NAGLU and SGSH, leads to bystander effects thus reducing the demand for gene transfer efficiency. The adeno-associated virus (AAV) vector system is one system with demonstrated therapeutic effect in a great variety of disease models. To date, no known pathogenesis has been linked to AAV in humans. Recombinant AAV (rAAV) vectors based on AAV serotype 2 (AAV2) have been used in numerous studies for neurological diseases, transducing both neuronal and non-neuronal cells in the CNS with demonstrated therapeutic benefits in treating MPS and other LSDs in animals and in patients with Parkinson's and Batten's disease. In the majority of rAAV-CNS gene therapy studies in LSDs, vectors were delivered by direct intracranial injection, which has limited potential for treating global CNS diseases. See, Sands et al., Acta Paediatr. Suppl., 97: 22-27 (2008); Fu et al., Mol. Ther., 5: 42-49 (2002); Cressant et al., J. Neurosci., 24: 10229-10239 (2004); Fraldi et al., Hum. Mol. Genet., 16: 2693-2702 (2007); Worgall et al., Hum. Gen. Ther., 19: 563-574 (2008) and Heldermon et al. Mol. Ther., 18: 873-880 (2010). To overcome these obstacles, more efficient delivery approaches have been developed with broad or global transduction, and functional benefits for the neurological disease in adult MPS IIIB mice. An intracisternal injection of rAAV2-hNAGLU vector in adult MPS IIIB mice, following mannitol pretreatment, led to deep periventricular transduction and clinical benefits. See Fu et al., J. Gene Med., 12: 624-633 (2010). Intravenous (IV) rAAV injection into neonatal MPS I, MPS VII and MPS IIIB mice led to long-term correction of lysosomal storage in both somatic and CNS tissues. See, Sands et al., Lab. Anim. Sci., 49: 328-330 (1999); Hartung et al., Mol. Ther., 9: 866-875 (2004) and Heldermon et al., supra. However, the BBB may still be permeable in neonatal mice while closed at birth in humans. Previously, in adult MPS IIIB mice, pretreatment with an N infusion of mannitol transiently disrupting the BBB facilitated the CNS entry of IV-delivered rAAV2, resulting in diffuse global CNS transduction and neurological correction. See, McCarty et al., Gene Ther., 16: 1340-1352 (2009).
  • Recombinant AAV9 vectors encoding the sulfamidase enzyme have been administered to MPSIIIA mice as reported in Ruzo et al., XVIII Annual Congress of the European Society of Gene and Cell Therapy: 1389 (Abstract Or 96) (October 2010) and Ruzo et al., Mol. Therap., 20(2): 254-266 (2012).
  • Adeno-associated virus (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). The nucleotide sequence of the AAV serotype 2 (AAV2) genome is presented in Srivastava et al., J Virol, 45: 555-564 (1983) as corrected by Ruffing et al., J Gen Virol, 75: 3385-3392 (1994). Cis-acting sequences directing viral DNA replication, encapsidation/packaging and host cell chromosome integration are contained within the ITRs. 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. Alternative splicing and non-consensus translational start sites are responsible for the production of the three related capsid proteins. A single consensus polyadenylation site is located at map position 95 of the AAV genome. The life cycle and genetics of AAV are reviewed in Muzyczka, Current Topics in Microbiology and Immunology, 158: 97-129 (1992).
  • 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. Moreover, AAV infects many mammalian cells allowing the possibility of targeting many different tissues in vivo. Moreover, 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). Furthermore, because 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, making cold preservation of AAV less critical. AAV may even be lyophilized. Finally, AAV-infected cells are not resistant to superinfection.
  • Multiple serotypes of AAV exist and offer varied tissue tropism. Known serotypes include, for example, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10 and AAV11. AAV9 is described in U.S. Pat. No. 7,198,951 and in Gao et al., J. Virol., 78: 6381-6388 (2004). 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): 321-328 (2005). The use of some serotypes of AAV to target cell types within the central nervous system, though, has required surgical intraparenchymal injection. See, Kaplitt et al., Lancet 369: 2097-2105 (2007); Marks et al., Lancet Neurol 7: 400-408 (2008); and Worgall et al., Hum Gene Ther (2008).
  • There remains a need in the art for products and methods for treating lysosomal storage disorders such as MPS IIIB and MPS IIIA.
  • SUMMARY
  • The present invention provides methods and materials useful for systemically delivering polynucleotides such as NAGLU polynucleotides or SGSH polynucleotides across the BBB.
  • According to the invention, gene delivery is achieved by utilizing, for example, AAV serotype 9 (AAV9). See, Foust et al., Nature Biotechnology, 27: 59-65 (2009); Duque et al., Mol. Ther. 17: 1187-1196 (2009); and Zincarelliet al., Mol. Ther., 16: 1073-1080 (2008). Vectors based on this serotype or functionally-related serotypes are able to cross the BBB unaided in neonate and adult animals. An added benefit to using AAV9 vectors is that pre-existing immunity is less common than for AAV2 serotype. The use of rh74 serotype AAV vectors among others is also contemplated by the invention.
  • In one aspect, the invention provides a method of delivering a NAGLU polynucleotide across the BBB comprising systemically administering a rAAV9 with a genome including the polynucleotide to a patient. In some embodiments the rAAV9 genome is a single-stranded genome.
  • More specifically, the present invention provides methods and materials useful for systemically delivering NAGLU polynucleotides across the blood brain barrier to the central and peripheral nervous system. In some embodiments, a method is provided of delivering a polynucleotide to the central nervous system comprising systemically administering a rAAV9 with a single-stranded genome including the genome to a patient. In some embodiments, a method of delivering a NAGLU polynucleotide to the peripheral nervous system comprising systemically administering a rAAV9 with a single-stranded genome including the polynucleotide to a patient is provided.
  • Even more specifically, in some embodiments, the NAGLU polynucleotide is delivered to brain. In some embodiments, the polynucleotide is delivered to the spinal cord. In some embodiments, the NAGLU polynucleotide is delivered to a lower motor neuron. In some embodiments, the polynucleotide is delivered to nerve and glial cells. In some embodiments, the glial cell is a microglial cell, an oligodendrocyte or an astrocyte. In some, embodiments, the rAAV9 is used to deliver a NAGLU polynucleotide to a Schwann cell.
  • Use of the NAGLU methods and materials is indicated, for example, for treating Sanfilippo syndrome Type B/MPS IIIB.
  • In another aspect, the invention provides a method of delivering an SGSH polynucleotide across the BBB comprising systemically administering a rAAV9 with a genome including the polynucleotide to a patient. In some embodiments, the rAAV9 genome is a self-complementary genome. In some embodiments the rAAV9 genome is a single-stranded genome.
  • More specifically, the present invention provides methods and materials useful for systemically delivering SGSH polynucleotides across the blood brain barrier to the central and peripheral nervous system. In some embodiments, a method is provided of delivering a polynucleotide to the central nervous system comprising systemically administering a rAAV9 with a self-complementary genome including the genome to a patient. In some embodiments, a method of delivering a SGSH polynucleotide to the peripheral nervous system comprising systemically administering a rAAV9 with a self-complementary genome including the polynucleotide to a patient is provided.
  • Even more specifically, in some embodiments, the SGSH polynucleotide is delivered to brain. In some embodiments, the polynucleotide is delivered to the spinal cord. In some embodiments, the SGSH polynucleotide is delivered to a lower motor neuron. In some embodiments, the polynucleotides is delivered to nerve and glial cells. In some embodiments, the glial cell is a microglial cell, an oligodendrocyte or an astrocyte. In some, embodiments, the rAAV9 is used to deliver a SGSH polynucleotide to a Schwann cell.
  • Use of the SGSH methods and materials is indicated, for example, for treating MPS IIIA.
  • In yet another aspect, administration of the rAAV9 encoding a NAGLU or SGSH polypeptide is preceded by administration of mannitol.
  • In still another aspect, the invention provides rAAV genomes comprising one or more AAV ITRs flanking a polynucleotide encoding a NAGLU. The NAGLU polynucleotide is operatively linked to transcriptional control DNAs, specifically promoter DNA and polyadenylation signal sequence DNA that are functional in target cells to form a gene cassette. The gene cassette may also include intron sequences to facilitate processing of an RNA transcript when expressed in mammalian cells.
  • In a further aspect, the invention provides rAAV genomes comprising one or more AAV ITRs flanking a polynucleotide encoding an SGSH. The SGSH polynucleotide is operatively linked to transcriptional control DNAs, specifically promoter DNA and polyadenylation signal sequence DNA that are functional in target cells to form a gene cassette. The gene cassette may also include intron sequences to facilitate processing of an RNA transcript when expressed in mammalian cells.
  • The rAAV genomes of the invention lack AAV rep and cap DNA. AAV DNA in the rAAV genomes (e.g., ITRs) 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. NC002077; the complete genome of AAV-2 is provided in GenBank Accession No. NC001401 and Srivastava et al., J. Virol., 45: 555-564 {1983); the complete genome of AAV-3 is provided in GenBank Accession No. NC1829; the complete genome of AAV-4 is provided in GenBank Accession No. NC001829; the AAV-5 genome is provided in GenBank Accession No. AF085716; the complete genome of AAV-6 is provided in GenBank Accession No. NC00 1862; at least portions of AAV-7 and AAV-8 genomes are provided in GenBank Accession Nos. AX753246 and AX753249, respectively; the AAV-9 genome is provided in Gao et al., J. Virol., 78: 6381-6388 (2004); the AAV-10 genome is provided in Mol. Ther., 13(1): 67-76 (2006); and the AAV-11 genome is provided in Virology, 330(2): 375-383 (2004).
  • NAGLU polypeptides contemplated include, but are not limited to, a NAGLU polypeptide with the amino acid sequence set out in SEQ ID NO: 2.
  • SGSH polypeptides contemplated include, but are not limited to, a SGSH polypeptide with the amino acid sequence set out in SEQ ID NO: 4.
  • The polypeptides contemplated include full-length proteins, precursors of full length proteins, biologically active subunits or fragments of full length proteins, as well as biologically active analogs (e.g., derivatives and variants) of any of these forms of polypeptides. Thus, polypeptides include, for example, those that (1) have an amino acid sequence that has greater than about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% or about 99% or greater amino acid sequence identity, over a region of at least about 25, about 50, about 100, about 200, about 300, about 400, or more amino acids, to a polypeptide encoded by a nucleic acid or an amino acid sequence described herein.
  • As used herein “biologically active derivative,” “biologically active fragment,” “biologically active analog” or “biologically active variant” includes any derivative or fragment or analog or variant of a molecule having substantially the same functional and/or biological properties of said molecule, such as enzymatic activities.
  • An “analog,” such as a “variant” or a “derivative,” is a compound substantially similar in structure to and having the same biological activity as, albeit in certain instances to a differing degree, a naturally-occurring molecule.
  • A “derivative,” for example, is a type of analog and refers to a polypeptide sharing the same or substantially similar structure as a reference polypeptide that has been modified, e.g., chemically.
  • A polypeptide variant, for example, is a type of analog and refers to a polypeptide sharing substantially similar structure and having the same biological activity as a reference polypeptide (i.e., “native polypeptide” or “native therapeutic protein”). Variants differ in the composition of their amino acid sequences compared to the naturally-occurring polypeptide from which the variant is derived, based on one or more mutations involving (i) deletion of one or more amino acid residues at one or more termini of the polypeptide and/or one or more internal regions of the naturally-occurring polypeptide sequence (e.g., fragments), (ii) insertion or addition of one or more amino acids at one or more termini (typically an “addition” or “fusion”) of the polypeptide and/or one or more internal regions (typically an “insertion”) of the naturally-occurring polypeptide sequence or (iii) substitution of one or more amino acids for other amino acids in the naturally-occurring polypeptide sequence.
  • Variant polypeptides include insertion variants, wherein one or more amino acid residues are added to a therapeutic protein amino acid sequence of the present disclosure. Insertions may be located at either or both termini of the protein, and/or may be positioned within internal regions of the therapeutic protein amino acid sequence. Insertion variants, with additional residues at either or both termini, include for example, fusion proteins and proteins including amino acid tags or other amino acid labels.
  • In deletion variants, one or more amino acid residues in a therapeutic protein polypeptide as described herein are removed. Deletions can be effected at one or both termini of the therapeutic protein polypeptide, and/or with removal of one or more residues within the therapeutic protein amino acid sequence. Deletion variants, therefore, include fragments of a polypeptide sequence.
  • In substitution variants, one or more amino acid residues of a therapeutic protein polypeptide are removed and replaced with alternative residues. In one aspect, the substitutions are conservative in nature and conservative substitutions of this type are well known in the art. Alternatively, the present disclosure embraces substitutions that are also non-conservative. Exemplary conservative substitutions are described in Lehninger, [Biochemistry, 2nd Edition; Worth Publishers, Inc., New York (1975), pp. 71-77] and are set out immediately below.
  • CONSERVATIVE SUBSTITUTIONS
    SIDE CHAIN
    CHARACTERISTIC AMINO ACID
    Non-polar (hydrophobic):
    A. Aliphatic A L I V P
    B. Aromatic F W
    C. Sulfur-containing M
    D. Borderline G
    Uncharged-polar:
    A. Hydroxyl S T Y
    B. Amides N Q
    C. Sulfhydryl C
    D. Borderline G
    Positively charged (basic) K R H
    Negatively charged (acidic) D E
  • Alternatively, exemplary conservative substitutions are set out immediately below.
  • CONSERVATIVE SUBSTITUTIONS II
    EXEMPLARY
    ORIGINAL RESIDUE SUBSTITUTION
    Ala (A) Val, Leu, Ile
    Arg (R) Lys, Gln, Asn
    Asn (N) Gln, His, Lys, Arg
    Asp (D) Glu
    Cys (C) Ser
    Gln (Q) Asn
    Glu (E) Asp
    His (H) Asn, Gln, Lys, Arg
    Ile (I) Leu, Val, Met, Ala, Phe,
    Leu (L) Ile, Val, Met, Ala, Phe
    Lys (K) Arg, Gln, Asn
    Met (M) Leu, Phe, Ile
    Phe (F) Leu, Val, Ile, Ala
    Pro (P) Gly
    Ser (S) Thr
    Thr (T) Ser
    Trp (W) Tyr
    Tyr (Y) Trp, Phe, Thr, Ser
    Val (V) Ile, Leu, Met, Phe, Ala
  • In yet further aspect, 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. Techniques to produce rAAV particles, in which an AAV genome to be packaged, rep and cap genes, and helper virus functions are provided to a cell are standard in the art. Production of rAAV requires that the following components are present within a single cell (denoted herein as a packaging cell): a rAAV genome, AAV rep and cap genes separate from (i.e., not in) the rAAV genome, and 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. Production of pseudotyped rAAV is disclosed in, for example, WO 01/83692 which is incorporated by reference herein in its 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. For example, 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. USA, 79:2077-2081), addition of synthetic linkers containing restriction endonuclease cleavage sites (Laughlin et al., 1983, Gene, 23:65-73) or by direct, blunt-end ligation (Senapathy & Carter, 1984, J. Biol. Chem., 259:4661-4666). The packaging cell line is then infected with a helper virus such as adenovirus. The advantages of this method are that the cells are selectable and are suitable for large-scale production of rAAV. Other examples of suitable methods employ adenovirus or baculovirus rather than plasmids to introduce rAAV genomes and/or rep and cap genes into packaging cells.
  • General principles of rAAV production are reviewed in, for example, Carter, 1992, Current Opinions in Biotechnology, 1533-539; and Muzyczka, 1992, Curr. Topics in Microbial. and Immunol., 158:97-129). Various approaches are described in Ratschin et al., Mol. Cell. Biol. 4:2072 (1984); Hermonat et al., Proc. Natl. Acad. Sci. USA, 81:6466 (1984); Tratschin et al., Mol. Cell. Biol. 5:3251 (1985); McLaughlin et al., J. Virol., 62:1963 (1988); and Lebkowski et al., 1988 Mol. Cell. Biol., 7:349 (1988). Samulski et al. (1989, J. Virol., 63:3822-3828); U.S. Pat. No. 5,173,414; WO 95/13365 and corresponding U.S. Pat. No. 5,658,776; WO 95/13392; WO 96/17947; PCT/US98/18600; WO 97/09441 (PCT/US96/14423); WO 97/08298 (PCT/US96/13872); WO 97/21825 (PCT/US96/20777); WO 97/06243 (PCT/FR96/01064); WO 99/11764; Perrin et al. (1995) Vaccine 13:1244-1250; Paul et al. (1993) Human Gene Therapy 4:609-615; Clark et al. (1996) Gene Therapy 3:1124-1132; U.S. Pat. No. 5,786,211; U.S. Pat. No. 5,871,982; and U.S. Pat. No. 6,258,595. The foregoing documents are hereby incorporated by reference in their entirety herein, with particular emphasis on those sections of the documents relating to rAAV production.
  • The invention thus provides packaging cells that produce infectious rAAV. In one embodiment packaging cells may be stably transformed cancer cells such as HeLa cells, 293 cells and PerC.6 cells (a cognate 293 line). In another embodiment, 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).
  • In another aspect, the invention provides rAAV (i.e., infectious encapsidated rAAV particles) comprising a rAAV genome of the invention. In some embodiments of the invention, the rAAV genome is a self-complementary genome.
  • The invention includes, but is not limited to, the exemplified rAAV named “rAAV9-CMV-hNAGLU.” The rAAV genome has in sequence an AAV2 ITR, the cytomegalovirus (CMV) immediate early promoter/enhancer, an SV40 intron (SD/SA), the NAGLU DNA set out in SEQ ID NO: 1, a polyadenylation signal sequence from bovine growth hormone and another AAV2 ITR. The DNA sequence of the vector genome is set out in SEQ ID NO: 5. The genome lacks AAV rep and cap DNA, that is, there is no AAV rep or cap DNA between the ITRs of the genome.
  • The invention also includes, but is not limited to, rAAV encoding SGSH. In some embodiments, the rAAV genome has in sequence an AAV2 ITR, the CMV immediate early promoter/enhancer, the SGSH DNA set out in SEQ ID NO: 3, a polyadenylation signal sequence from bovine growth hormone and a AAV2 ITR lacking the terminal resolution site. In some embodiments, the rAAV genome has in sequence an AAV2 ITR, the mouse U1a promoter, the SGSH DNA set out in SEQ ID NO: 3, a polyadenylation signal sequence from bovine growth hormone and a AAV2 ITR lacking the terminal resolution site. In some embodiments, rAAV genome has in sequence an AAV2 ITR, the mouse U1a promoter, an intron, the SGSH DNA set out in SEQ ID NO: 3, a polyadenylation signal sequence from bovine growth hormone and a AAV2 ITR lacking the terminal resolution site. The genomes lack AAV rep and cap DNA, that is, there is no AAV rep or cap DNA between the ITRs of the genomes.
  • NAGLU and SGSH DNAs include, without limitation, those that (1) hybridize under stringent hybridization conditions to a nucleic acid encoding an amino acid sequence as described herein, and conservatively modified variants thereof; (2) have a nucleic acid sequence that has greater than about 95%, about 96%, about 97%, about 98%, about 99%, or higher nucleotide sequence identity, over a region of at least about 25, about 50, about 100, about 150, about 200, about 250, about 500, about 1000, or more nucleotides (up to the full length sequence of the mature protein), to a nucleic acid sequence as described herein. Exemplary “stringent hybridization” conditions include hybridization at 42° C. in 50% formamide, 5×SSC, 20 mM Na.PO4, pH 6.8; and washing in 1×SSC at 55° C. for 30 minutes. It is understood that variation in these exemplary conditions can be made based on the length and GC nucleotide content of the sequences to be hybridized. Formulas standard in the art are appropriate for determining appropriate hybridization conditions. See Sambrook et al., Molecular Cloning: A Laboratory Manual (Second ed., Cold Spring Harbor Laboratory Press, 1989) §§9.47-9.51.
  • The rAAV 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.
  • In an additional aspect, the invention contemplates compositions comprising rAAV of the present invention encoding an NAGLU polypeptide. These compositions may be used to treat mucopolysaccharidosis IIIB. In other embodiments, compositions of the present invention may include two or more rAAV encoding different polypeptides of interest.
  • In still an additional aspect, the invention contemplates compositions comprising rAAV of the present invention encoding an SGSH polypeptide. These compositions may be used to treat mucopolysaccharidosis IIIA. In other embodiments, compositions of the present invention may include two or more rAAV encoding different polypeptides of interest.
  • 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 counterions such as sodium; and/or nonionic surfactants such as Tween, pluronics or polyethylene glycol (PEG).
  • 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 1×106, about 1×107, about 1×108, about 1×109, about 1×1010, about 1×1011, about 1×1012, about 1×1013 to about 1×1014 or more DNase resistant particles (DRP) per ml. 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 1×1011 vg/kg, about 1×1012, about 1×1013, about 1×1014, about 1×1015, about 1×1016 or more viral genomes per kilogram body weight in an adult. For a neonate, the dosages of rAAV may range from about 1×1011, about 1×1012, about 3×1012, about 1×1013, about 3×1013, about 1×1014, about 3×1014, about 1×1015, about 3×1015, about 1×1016, about 3×1016 or more viral genomes per kilogram body weight.
  • Treatment by methods of the invention comprises the step of administering an intravenous (IV) effective dose, or effective multiple doses, of a composition comprising a rAAV of the invention to an animal (including a human being) in need thereof. If the dose is administered prior to development of a disorder/disease, the administration is prophylactic. If the dose is administered after the development of a disorder/disease, the administration is therapeutic. In embodiments of the invention, 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.
  • 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., transient or long-term immunosuppression) are specifically contemplated, as are combinations with novel therapies.
  • Compositions suitable for systemic (IV) use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating actions of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of a dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal and the like. In many cases it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by use of agents delaying absorption, for example, aluminum monostearate and gelatin, and Tween family of products (e.g., Tween 20).
  • Sterile injectable solutions are prepared by incorporating rAAV in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filter sterilization. Generally, dispersions are prepared by incorporating the sterilized active ingredient into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze drying technique that yield a powder of the active ingredient plus any additional desired ingredient from the previously sterile-filtered solution thereof.
  • Transduction of cells with rAAV of the invention results in sustained expression of NAGLU or SGSH polypeptide. Transduction may be carried out with gene cassettes comprising tissue specific control elements, for example, promoters that allow expression specifically within neurons or specifically within astrocytes. Examples include neuron specific enolase and glial fibrillary acidic protein promoters. Inducible promoters under the control of an ingested drug may also be developed.
  • It will be understood by one of ordinary skill in the art that a polynucleotide delivered using the materials and methods of the invention can be placed under regulatory control using systems known in the art. By way of non-limiting example, it is understood that systems such as the tetracycline (TET on/off) system [see, for example, Urlinger et al., Proc. Natl. Acad. Sci. USA 97(14):7963-7968 (2000) for recent improvements to the TET system] and Ecdysone receptor regulatable system [Palli et al., Eur J. Biochem 270: 1308-1315 (2003] may be utilized to provide inducible polynucleotide expression.
  • Thus, the invention provides methods of systemically administering an effective dose (or doses, administered essentially simultaneously or doses given at intervals) of rAAV of the invention to a patient in need thereof.
  • BRIEF DESCRIPTION OF THE DRAWING
  • FIG. 1 is a map of the rAAV-CMV-hNAGLU vector genome.
  • FIG. 2 shows improved behavior and extended survival in MPS IIIB mice after systemic gene transfer by rAAV-CMV-hNAGLU. FIG. 2 a. Hidden task in water maze (n=11/group). Day 1: test trial. FIG. 2 b. Latency to fall from a rotarod (n=11/group). FIG. 2 c. Survival (i 5/group, P<0.001). +/+: wt; −/−: MPS IIIB; AAV9-L, AAV9-H: MPS IIIB mice treated with 5×1012 or 1.5×1013 vg/kg rAAV9-hNAGLU vector, respectively. *: P<0.05 (vs. +/+); #: P<0.05 (vs. AAV9-L); ̂: P<0.05 (vs. AAV9-H); &: P>0.05 (vs. −/−). Repeated measures ANOVA analyses: @: day effect P<0.01; $: group (treatment effect) P<0.01; %: day-group interaction P41.020 (rotarod).
  • FIG. 3 shows rAAV9-mediated expression of functional rNAGLU in tissues. Tissues from MPS IIIB mice treated with rAAV9-hNAGLU were assayed for NAGLU activity (6 and 9 mo pi) (n=5-6/group). FIG. 3 a. Dose-response. +/+: wt; AAV9-11, AAV9-L: MPS IIIB mice treated with 1.5×1013(AAV9-H) or 5×1012 vg/kg (AAV9-L) vector; FIG. 3 b. Impact of mannitol pretreatment. M+/M−: MPS IIIB mice treated with 2×1013 vg/kg vector with (M+) or without (M−) mannitol pretreatment. FIG. 3 c. Plasma NAGLU activity (n=3-4). +/−: heterozygotes. No significant difference in tissue NAGLU activity was detected at 6 and 9 months pi. Data shown are means±SD of combined data on tissues from mice at 6 and 9 mo pi. *: P<0.01 vs. +/+; #: P<0.05 vs. AAV9-H or M+: P>0.05 vs. +/+. @: P<0.05 vs. +/−.
  • FIG. 4 shows the significant reduction of GAG content in the CNS and somatic tissues. Tissues from MPS IIIB mice treated with rAAV9-hNAGLU were assayed to quantify GAG content (6 and 9 mo pi). FIG. 4 a. Dose response. FIG. 4 b. Impact of mannitol pretreatment. +/Ai wt; −/−: MPS IIIB; AAV9-H, AAV9-L: MPS MB mice treated with 1.5×1013 vg or 5×1012 vg/kg vector; M+, M−: MPS IIIB mice treated with rAAV9 vector (2×1013 vg/kg) with or without mannitol pretreatment. Data shown are means±SD (n=5-6), combining data from tissues collected at 6 and 9 mo pi. *: P<0.01 vs. +/+; #: P<0.05 vs. AAV9-H or M+; ̂: P<0.05 vs. AAV9-L or M−; +: P>0.05 vs. +/+.
  • FIG. 5 shows rAAV9-mediated correction of astrocytosis and neurodegeneration in MPS IIIB mice. Brain sections of MPS IIIB mice treated with rAAV9-CMV-hNAGLU vector (6 mo pi) were assayed for GFAP by immunofluorescence and stained with toluidine blue for histopathology. FIG. 5 a. Number of astrocytes: Data are means±SD of GFAP+ cells per 330×433 pm on 6-8 IF-GFAP-staining sections/mouse, from 3 mice/group. FIG. 5 b. Number of purkinje cells: Data are means±SD of purkinje cells/200 p.m (in length) in ansiform lobules in cerebellum on 6 toluidine blue stained sections/mouse, from 3 mice/group. NT: non-treated MPS IIIB mouse; AAV9: MPS IIIB mouse treated with rAAV9. CTX: cerebral cortex; ST: Striatum; TH: thalamus; BS: Brain stem. *: P<0.01 vs. non-treated.
  • FIG. 6 shows rAAV9-mediated expression of functional rSGSH in tissues of treated MPSIIIA mice. For each tissue, AAV9, rh74 and untreated result bars are respectively shown from left to right.
  • FIG. 7 shows a significant reduction of GAG content in tissues of treated MPSIIIA mice. For each tissue, AAV9, rh74 and untreated result bars are respectively shown from left to right.
  • FIG. 8 shows an improvement in cognitive behavior assays after treatment of one-month old MPSIIIA mice with low dose scAAV9 or rh74-U1a-SGSH. In the lower graphs, for each tissue, untreated, wild type and either AAV9 or rh74 result bars are respectively shown from left to right.
  • FIG. 9 shows a significant reduction of GAG content in tissues of MPSIIIA mice treated at 2 or 6 months of age. For each tissue, wild type, untreated, two-month and six-month result bars are respectively shown from left to right.
  • FIG. 10 shows an improvement in cognitive behavior assays after treatment of two- or six-month old MPSIIIA mice with high dose scAAV9 or rh74-U1a-SGSH. In the lower graphs, for each tissue, untreated, wild type and either AAV9 or rh74 result bars are respectively shown from left to right.
  • DETAILED DESCRIPTION
  • The present invention is illustrated by the following examples relating to delivery of human NAGLU (hNAGLU) genes to the spinal cord via intravenous delivery of rAAV9. Example 1 describes rAAV encoding hNAGLU. Example 2 describes the administration of the rAAV encoding hNAGLU to MPSIIIB mice. Examples 3 through 6 describe the beneficial results of administration of the rAAV. Example 7 discusses the significance of the results. Example 8 describes rAAV encoding SGSH. Examples 9 through 11 describe administration of various dosages of rAAV encoding SGSH to MPSIIIA mice of varying ages, as well as the beneficial effects of the administration.
  • EXAMPLE 1 Recombinant AAV (rAAV) Viral Vectors Encoding NAGLU
  • A rAAV vector plasmid, containing AAV2 ITRs, an immediate early CMV promoter/enhancer, an SV40 intron, a human α-N-acetylglucosaminidase coding region, a bGH polyadenylation signal sequence, and ampicillin resistance gene, was used to produce a rAAV9-CMV-hNAGLU viral vector.
  • Recombinant AAV9 viral vectors with the hNAGLU-encoding genome were produced in 293 cells using three-plasmid co-transfection, and purified as described in Zolotukhin et al., Gene Ther., 6: 973-985 (1999). This vector is referred to as “rAAV9-CMV-hNAGLU” herein. The vector genomes contained minimal elements required for transgene expression, including AAV2 terminal repeats, a human cytomegalovirus (CMV) immediate-early promoter, SV40 splice donor/acceptor signal, a human NAGLU coding sequence (SEQ ID NO: 1), and bGH polyadenylation signal. SEQ ID NO: 5 is the DNA sequence of the vector genome. FIG. 1 is a map of the vector genome wherein the length of the various elements of the genome is indicated below the element.
  • A control self-complementary AAV encoding green fluorescent protein, scAAV9-CMV-GFP was also produced, containing AAV2 terminal repeats, a human cytomegalovirus (CMV) immediate-early promoter, SV40 splice donor/acceptor signal, a eGFP coding sequence, and SV40 polyadenylation signal.
  • EXAMPLE 2 Administration of Viral Vectors
  • An MPS IIB3 knock-out mouse colony [Li et al., Proc. Natl. Acad. Sci. USA, 96: 14505-14510 (1999) was maintained on an inbred background (C57BL/6) of backcrosses of heterozygotes. All care and procedures were in accordance with the Guide for the Care and Use of Laboratory Animals [DHHS Publication No. (NIH) 85-23]. The genotypes of progeny mice were identified by PCR.
  • To assess the therapeutic efficacy of rAAV9 gene delivery, 4-6-week-old MPS IIIB mice were treated with an IV injection of rAAV9-CMV-hNAGLU (5×1012 or 1.5×1013 vg/kg, n=11/group). Separately, other MPS IIIB mice were treated with 2×1013 vg/kg rAAV9-CMV-hNAGLU, with or without mannitol pretreatment (n=5/group), to assess the impact on CNS entry. Controls were sham-treated (phosphate-buffered saline) wild type (wt) and MPS IIIB littermates (n=11). Tissue analyses were carried out at 6 months and 9 months (n=2-4/group) post-injection (pi).
  • Additionally, self-complementary AAV (scAAV) vector carrying a cytomegalovirus-green fluorescent protein (CMV-GFP) transgene (5×1012 vg/kg) was injected IV into 6-8-week-old wt mice (n=4/group) to determine the distribution of transgene expression (1 month pi), as a comparison to rAAV9-hNaGlu treatment.
  • Results are presented below.
  • EXAMPLE 3 Behavioral Tests
  • The rAAV9-CMV-hNaGlu-treated MPS BIB mice and controls were tested for behavioral performance at approximately 5.0-5.5 months of age as follows.
  • Hidden Task in the Morris Water Maze
  • A water maze (diameter=122 cm) was filled with water (45 cm deep, 24-26° C.) containing 1% white TEMPERA paint, located in a room with numerous visual cues. See Warburton et al., J. Neurosci, 21: 7323-7330 (2001). Mice were tested for their ability to find a hidden escape platform (20×20 cm) 0.5 cm under the water surface. Each animal was given four trials per day, across three days, as described previously. Measures were taken of latency to fmd the platform (sec) via an automated tracking system (San Diego Instruments). Results are shown in FIG. 2 a.
  • Rotarod
  • Mice were tested on an accelerating rotarod (Med Associate, Inc.) to assess motor coordination. See Lijam et al., Cell, 90895-905 (1997). Rotation speed was set at an initial value of 3 revolutions per minute (rpm), with a progressive increase to a maximum of 30 rpm across five minutes (the maximum trial length). For the first test session, animals were given three trials, with 45 seconds between each trial. Two additional trials were given 48 hours later. Measures were taken for latency to fall from the top of the rotating barrel. Results are shown in FIG. 2 b.
  • Statistical Analyses
  • Means, standard deviation (SD) and unpaired student t-test were used to analyze quantitative data. Behavioral measures were taken by an observer blind to experimental treatment. Behaviroal testing data were also analyzed using repeated measures ANOVA (SAS 9.1.3) to determine the significance of the variances among treatment and control groups and testing days.
  • Results of Behavioral Tests
  • All mice treated IV with 5×1012 or 1.5×1013 vg/kg rAAV9-NAGLU were tested for behavior at 5-5.5 mo of age to assess the neurological impacts. Both dosage groups exhibited significant decreases in latency to find a hidden platform in a water maze (FIG. 2 a), and significantly longer latency to fall from an accelerating rotarod (FIG. 2 b), compared with non-treated MPS IIIB mice, indicating the correction of cognitive and motor function. There were no significant differences in behavior performance between these two dose groups.
  • EXAMPLE 4 Longevity Assessment
  • Following the rAAV9-hNaGlu vector injection(s), mice were continuously observed for the development of endpoint symptoms, or until death occurred. The endpoint was when the symptoms of late stage clinical manifestation (urine retention, rectal prolapse, protruding penis) in MPS IIIB mice became irreversible, or when wt control mice were 24 months or older. Longevity data were analyzed using Kaplan-Meier method. The significance level was set at P<0.05. Results are shown in FIG. 2 c.
  • Ten rAAV9-treated MPS IIIB mice, five from each dose group, were observed for longevity. All ten survived >16.9 months (with one mouse of the low-dose group dying at age of 16.1 months) and the majority of them survived 18.9-27.4 months within the normal range of lifespan, while all non-treated MPS IIIB mice died at 8-12 months of age (P<0.001) (FIG. 2 c). These data demonstrate that a single IV rAAV9 vector injection alone is functionally beneficial in treating the CNS disease and increasing longevity in MPS IIIB mice.
  • EXAMPLE 5 Tissue Analyses
  • In the therapeutic studies above, tissue analyses were carried out at 6 mo and 9 mo post injection (pi). Mice were anesthetized with 2.5% Avertin before tissue collection. Brain, spinal cord and multiple somatic tissues were collected on dry ice or embedded in OCT compound and stored at −70° C., before being processed for analyses. Tissues were also processed for paraffm sectioning.
  • Tissue samples from scAAV9-GFP vector-treated mice were collected for analysis 4-5 weeks pi. The mice were anesthetized with 2.5% Avertin and then perfused transcardially with cold PBS (0.1M, pH7.4), followed by 4% paraformaldehyde in phosphate buffer (0.1M, pH7.4). The entire brain and spinal cord, as well as multiple somatic tissues (including liver, kidney, spleen, heart, lung, intestine and skeletal muscles), were collected and fixed in 4% paraformaldehyde overnight at 4° C. before being further processed for vibratome sectioning.
  • NAGLU Activity Assay
  • Tissues were analyzed at 6 mo and/or 9 mo pi by NAGLU activity assay to determine the distribution and level of rAAV9-mediated transgene expression. Tissue samples were assayed for NaGlu enzyme activity following a published procedure with modification. The assay measures 4-methylumbelliferone (4MU), a fluorescent product formed by hydrolysis of the substrate 4-methylumbellireyl-N-acetyla-D-glucosaminide. The NaGlu activity is expressed as unit/mg protein. 1 unit is equal to 1 nmol 4MU released/h at 37° C. Results are shown in FIG. 3.
  • GAG Content Measurement
  • GAG was extracted from tissues following published procedures [van de Lest et al., Anal. Biochem. 221: 356-361(1994)] with modification [Fu et al., Gene Ther., 14: 1065-1077 (2007). Dimethylmethylene blue (DMB) assay was used to measure GAG content [de Jong et al., Clin. Chem., 35: 1472-1477 (1989)]. The GAG samples (from 0.5-1.0 mg tissue) were mixed with H2O to 40 ml before adding 35 nM DMB (Polysciences CEO 03610-1) in 0.2 mM sodium formate buffer (SFB, pH 3.5). The product was measured using a spectrophotometer (0D535). The GAG content was expressed as μg/mg tissue. Urine GAG content was also measured. Heparan sulfate (Sigma, H9637) was used as standard. Results are shown in FIG. 4.
  • Immunofluorescence
  • Immunofluorescence (IF) was performed to identify cells expressing hNAGLU, GFP or glial fibrillary acidic protein (GFAP) for astrocytes, using antibodies against hNaGlu (a kind gift from Dr. EF Neufeld, UCLA), GFP (Invitrogen) or GFAP (Chemicon), and corresponding secondary antibody conjugated with AlexaFluor568 or AlexaFluor488 (Molecular Probes). The IF staining was performed on thin cryostat sections (8 p.m) of tissue samples following procedures recommended by the manufacturers. The sections were visualized under a fluorescence microscope.
  • Histopathology
  • Tissues were assayed for histopathology to visualize the impact of IV rAAV9-NAGLU gene delivery on the lysosomal storage pathology in MPS IIIB mice. Histopathology was performed following standard methods. Paraffm sections (41.un) were fixed with 4% paraformaldehyde in phosphate buffer (0.1 M, pH 7.2) at 4° C. for 15 min and stained with 1% toluidine blue at 37° C. for 30 min to visualize lysosomal GAG. The sections were mounted, and imaged under a light microscope.
  • Quantitative Real Time PCR
  • Total DNA was isolated from tissue samples of treated and nontreated MPS IIIB mice using Qiagen DNeasy columns. Brain DNA was isolated from midbrain tissue. The DNA samples were analyzed by quatitative real-time PCR, using Absolute Blue QPCR Mix (Thermo Scientific, Waltham, Mass.) and Applied Biosystems 7000 Real-Time PCR System, following the procedures recommended by the manufacturer. Taqman primers specific for the CMV promoter were used to detect rAAV vector genomes: f: GGCAGTACATC AAGTGTATC (SEQ ID NO: 6); r: ACCAATGG TAATAG CGATGAC (SEQ ID NO: 7); probe: [6˜FAM]AATGACGGTAAAT GGCCCGC[TAMRA˜6˜FAM] (SEQ ID NO: 8). Genomic DNA was quantified in parallel samples using β-actin specific primers: f: GTCATCAC TATTG GCAACGA (SEQ ID NO: 9); r: CTCAGGAGTTTTGTCACCTT (SEQ ID NO: 10); probe: [6˜FAM]TTCCGATGCCCT GAGGCTCT[Tamra˜Q] (SEQ ID NO: 11). Genomic DNA from nontreated MPS IIIB mouse tissues was used as controls or background and absence of contamination. Global CNS and widespread somatic restoration of NAGLU.
  • Tissue Analysis Results
  • Tissues were analyzed at 6 months and/or 9 months pi by immunofluorescence (IF) and NAGLU activity assay to determine the distribution and level of rAAV9-mediated transgene expression. NAGLU-specific IF was detected throughout the brains of treated mice, in neurons, glia, and abundant endothelial cells in capillaries and larger blood vessels, in an apparently dose—dependent fashion. No significant differences were observed in the distribution or levels of rNaGlu signal between 6 months and 9 months pi. NAGLU-positive glial cells were not costained with anti-glial fibrillary acidic protein (GFAP) Ab, and were likely to be oligodendrocytes, based on their morphology. Importantly, while rNAGLU IF was observed in the brains of all rAAV9-treated mice, mannitol pretreatment did appear to increase the number of transduced cells in the CNS.
  • Differential transduction levels were observed in peripheral organs. The rNAGLU protein was detected in 20-40% of hepatocytes, >95% of cardiomyocytes, and 10-30% of skeletal myocytes. The distribution of rAAV9-transduced hepatocytes was uniform throughout the liver. Transduction in abundant neurons in myenteric plexus and submucosal plexus of the intestine was observed, suggesting efficient targeting of the peripheral nervous system (PNS). The rNAGLU signals were mostly present in granules, whereas scAAV9-mediated GFP signals were uniform in the cytoplasm of transduced cells, suggesting correct lysosomal trafficking of rNAGLU. Transduction of endothelial cells was also observed in peripheral tissues of rAAV9-GFP vector-treated mice.
  • EXAMPLE 6 Enzyme Function Assays
  • Function of the recombinant NAGLU and resulting effects in animals were also analyzed in the therapeutic studies above.
  • rNAGLU Enzymatic Function
  • Transgene enzymatic activity was assayed to quantify the expression and the functionality of rAAV9-mediated rNAGLU. There were no significant differences in tissue NAGLU activity at 6 and 9 months pi, suggesting stable transduction. The rAAV mediated rNaGlu was metabolically functional and the tissue rNAGLU activity was dose-dependent, with approximately normal levels in the brains of mice receiving 5×1012 vg/kg vector, and supra-physiologic levels in the brains of mice receiving 1.5×1013 vg/kg (FIG. 3 a). In both dose groups, we detected NAGLU activity at normal or subnormal levels in the liver, lung and intestine (FIG. 3 a), supra-physiologic levels in the skeletal muscles (FIG. 3 a) and heart (40 & 100 units/mg protein, data not shown), and low levels in the spleen, but no detectable NAGLU activity in the kidney. A low level of NAGLU activity was detected in the kidneys of the mice treated with 2×1013 vg/kg vector (FIG. 3 b). Mannitol pretreatment led to an increase in NAGLU activity in the brain (though not significant due to high individual variation), liver, spleen, lung and intestine, but a decrease in the heart and skeletal muscle (FIG. 3 b). No detectable NAGLU activity (<0.03 unit/mg) was observed in tissues from non-treated MPS IIIB mice.
  • rNaGlu Secretion
  • Plasma samples were assayed for NAGLU activity to assess the secretion of the enzyme. Activity was detected in the plasma of all rAAV9-treated MPS MB mice at or near heterozygote levels, though lower than homozygous wt levels (FIG. 3 c). Mannitol pretreatment resulted in significant reduction in plasma NAGLU activity (FIG. 3 c). These data indicate that the rNAGLU was secreted, though the source tissue or cell type is not clear.
  • GAG Content Reduction
  • Tissues were assayed for GAG content to quantify the impact of IV rAAV9-NAGLU gene delivery on the lysosomal storage pathology in MPS IIIB mice. The single IV rAAV9-NAGLU injection led to a reduction of GAG content to normal levels in the brain, liver, heart, lung, intestine and skeletal muscle in mice of all four treatment groups (FIG. 4). Doses of 5×1012 μg or 1.5×1013 vg/kg resulted in partial GAG reduction in the spleen but had no impact in kidney (FIG. 4 a).
  • Treatment with 2×1013 vg/kg led to a decrease of GAG to normal levels in the spleen, and partial GAG reduction in the kidney (FIG. 4 b), consistent with the observed enzyme activity levels.
  • Histopathology Correction
  • Histopathology showed complete clearance or reduction of lysosomal storage lesions in the vast majority of CNS areas, including cerebral cortex, thalamus, brain stem, hippocampus, and spinal cord in all four treatment groups. There were decreases in the size, number of vacuoles, and number of cells with lysosomal storage lesions, even in the few brain areas that did not show a complete correction, such as purkinje cells and cells in the striatum and hypothalamus. Importantly, the majority of brain and spinal cord parenchymal cells exhibited a well defined normalized morphology. Immunofluorecence detection for the lysosomal marker, LAMP-1, showed that IV infusion of rAAV9-NAGLU vector at all doses also resulted in marked reduction of LAMP-1 signal, especially in neurons, throughout the brain. This further supports the conclusion that the amount of vector crossing the BBB was sufficient for efficient correction of CNS lysosomal storage pathology.
  • In somatic tissues, complete clearance of lysosomal storage lesions in the livers of all rAAV9-hNaGlu treated mice was observed as well as attenuation of nuclear shrinkage, a marker of cell stress and damage.
  • Correction of Gliosis and Neurodegeneration
  • In order to determine whether the rAAV9-hNaGlu vector delivery had an impact on astrocytosis, a major secondary neuropathology of MPS IIIB, brain sections were assayed by immunofluorecence for GFAP expressing cells. Significant decreases in astrocyte numbers in gray matter throughout the brain of treated mice were observed compared to untreated at 6 mo and 9 mo pi (FIG. 5 a). Histopathology also revealed significant increases in the numbers of neurons, such as Purkinje cells (FIG. 5 b), in the brains of treated MPS IIIB mice. These data strongly indicate the amelioration of astrocytosis and neurodegeneration, which are hallmarks of secondary neuropathologies in MPS IIIB, in response to the rAAV9 treatment.
  • Vector Genome Distribution
  • Quantitative real-time PCR was performed to compare the amount of rAAV9-CMV-hNaglu vector entering the CNS versus somatic tissues. Table 1 shows the distribution of the vector genome in different tissues/organs of MPS IIIB mice treated with IV vector injection at varying doses. The highest concentrations of vector genome were detected in liver (8.20±4.73-32.09±3.93 copies/cell), followed by heart (0.07-0.22 copies/cell), and brain (0.06±0.001-0.15±0.02 copies/cell), and very low copy numbers were detected in other tissues/organs (Table 1). This differential vector distribution in rAAV9-treated MPS IIIB mice largely correlated with the distribution of rNAGLU IF and enzymatic activity. Notably, mannitol pretreatment increased the vector copy numbers in the brain, correlating with brain NAGLU activity levels. Furthermore, these data reflect persistence of vector genome distribution in treated mice at 6 months pi, supporting a stable long-term transduction. Levels of vector genome copies correlating with rNAGLU activity and distribution were not detectable, possibly due to difficulties in quantitative isolation of DNA from muscle tissue.
  • TABLE 1
    Estimated vector genome in the liver and brain of rAAV9-treated mice
    Vector genome (copy/cell)
    Mice n Liver Brain Heart
    rAAV9-L 2  8.20 ± 4.73 0.07 ± 0.07 0.07*
    rAAV9-H 3 10.86 ± 2.94  0.09-10.47 0.13 ± 0.07
    rAAV9-M− 2 21.97 ± 6.43  0.06 ± 0.001 0.22*
    rAAV9-M+ 3 32.09 ± 3.93 0.15 ± 0.02 0.14*
    Non-treated 1 0.000 0.000 0.00 
    Mouse tissue samples (6mo pi) were assayed in duplicates for vector genome copy numbers by qPCR.
    Data is expressed as vector copy/cell (means ± SD).
    rAAV9-L: IV infusion of 5 × 1012 vg/kg;
    rAAV9-H: IV infusion of 1.5 × 1013 vg/kg;
    rAAV9-M−: IV infusion of 2 × 1013 vg/kg without mannitol pretreatment;
    rAAV9-M+: IV infusion of 2 × 1013 vg/kg following mannitol pretreatment.
    *Data from 1 sample in duplicates.
  • EXAMPLE 7 Discussion
  • This study demonstrates the first significant therapeutic benefit for treating MPS IIIB in adult animals from systemic gene delivery to the CNS without additional treatment to disrupt the BBB. A single IV injection of hNAGLU-expressing rAAV9 vector was sufficient to significantly improve cognitive and motor functions, and greatly prolong survival in MPS IIIB mice. In the present study using rAAV9, the increased longevity exceeds the outcome of previous studies using rAAV2 vector delivered through either intracisternal injection, or systemic injection following mannitol pretreatment. The rNAGLU enzyme was clearly secreted and functional, leading to a significant bystander effect, and efficient degradation of heparan sulfate GAG in CNS tissues. Importantly, the clinically meaningful therapeutic benefits of the IV-delivered rAAV9 vector in MPS IIIB mice were achieved at a lower dose than the mannitol-facilitated, systemically delivered rAAV2 vector. The enhanced rAAV9-CNS transduction in response to mannitol pretreatment suggests further potential for reducing the vector dose, and the attendant risk and burden to patients.
  • The IV vector injection resulted in a ubiquitously diffuse, global rAAV9-NaGlu transduction throughout the CNS, reflecting the expected distribution pattern for vascular delivery. This contrasts sharply with the focal gradient distribution typically achieved through direct brain parenchymal injection, or the periventricular transduction pattern from intracisternal and intraventricular injection. While similar to the pattern of transgene expression from IV-delivered rAAV2 after mannitol pretreatment, the extent of rAAV9 transduction was significantly higher in all areas of the brain. This correlates with the increased effects on longevity and cognitive function compared to that previously achieved using rAAV2-mannitol treatment, and the normal or above normal levels of NAGLU activity in the CNS. These findings strongly support the use of the trans-BBB neurotropic rAAV9 as a vector for CNS gene therapy and reinforce the view that efficient CNS delivery is the most critical issue for developing therapies to treat MPS IIIB.
  • The rAAV9-transduced CNS cells include neurons, glia and endothelia. Neuronal cell transduction appears to be non-preferential, including most types of neurons throughout the brain. In contrast, the transduction of glial cells appears to be cell-type specific, targeting predominantly oligodendrocyte-like cells, though it is unclear whether this is a receptor- or promoter-specific phenomenon. In a previous report [Faust et al., supra] describing predominant transduction of astocytes after systemic injection of rAAV9 vector in adult mice, a hybrid chicken J3-actin/CMV-enhancer promoter was used, rather than the CMV enhancer-promoter used in the present study.
  • In normal cells, 5-20% of newly synthesized lysosomal protein is secreted and available to be taken up by neighboring cells, leading to the by-stander effect. The widespread clearance/reduction of lysosomal storage pathology, and normalized tissue GAG content, strongly support an efficient by-stander effect from the rAAV9
    Figure US20130039888A1-20130214-P00001
    mediated rNAGLU. The abundant transduction of endothelial cells in the brain may be an important contributor to the effectiveness of rAAV9 gene delivery for MPS IIIB because of the close association between CNS cells and brain microvascular endothelial cells, which together constitute the neurovascular unit. While the observed high levels of rNAGLU expression stem from the transduction of a relatively small number of CNS cells, it is sufficient to correct the neuropathology leading to functional correction of the neurological disorders.
  • The rAAV9 treatment also led to a regular morphology in CNS cells, and the correction of major secondary neuropathology, astrocytosis, and neurodegeneration. It is worth noting that this level of correction of CNS pathology was not achieved in previous studies using rAAV2-hNAGLU vector with mannitol. While neuropathology is the primary cause of mortality in MPS IIIB patients, somatic correction may provide additional therapeutic benefits, since lysosomal storage pathology inevitably manifests in virtually all organs. The IV-delivered rAAV9 exhibited broad tropism in peripheral tissues in a distinct pattern, as previously reported, reflecting extensive extravasation and cell-type specific transduction. This led to complete, long
    Figure US20130039888A1-20130214-P00001
    term correction of lysosomal storage in multiple somatic tissues even at a relatively low dose. Again, relatively low levels of transduction in some tissues were associated with clearance of lysosomal storage of GAGs in the organs, consistent with a significant contribution from the by-stander effect of secreted rNAGLU enzyme. It is not clear whether the by-stander correction in peripheral tissues is mediated by enzyme secreted from neighboring cells within the same tissue, or circulating rNAGLU secreted by more extensively transduced tissues, in a manner analogous to enzyme replacement therapy. However, the observation of partial GAG reduction in the kidney only at the highest vector dose, correlating with detectable transduction in the kidney only at that dose, suggests that the by-stander effect may be primarily local in this tissue. The primary source of circulating NAGLU may be liver, muscle, or endothelium. However, the decrease in plasma levels in response to mannitol pretreatment correlated with decreased transduction in muscle rather than liver, suggesting that liver may not be the primary source.
  • Another important observation is the efficient transduction of neurons in myenteric plexus and submucosal plexus of the intestine, potentially enabling correction of not only the CNS but also the PNS at all levels via systemic delivery. This suggests that neurotropism is a general property of the AAV9 serotype, and not dependent on the specific structure of the brain neurovascular unit. Broad neurotropism is a valuable property in gene therapy for the treatment of MPS IIIB, considering that lysosomal storage pathology manifests not only in the CNS but also in the PNS.
  • EXAMPLE 8 Recombinant AAV (rAAV) Viral Vectors Encoding SGSH
  • A rAAV vector plasmid was used to produce three different rAAV9-CMV-hSGSH viral vectors.
  • The three self-complementary AAV hSGSH vector-producing plasmids were constructed using conventional plasmid cloning techniques. Each vector genome contains an SGSH coding region (SEQ ID NO: 3) and either the mouse U1a promoter, with or without an intron, or a CMV promoter without an intron, Each vector genome also contains a bGH polyadenylation signal. Each self-complementary vector plasmid construct contains one intact AA2 terminal repeat and one modified AAV2 terminal repeat missing the terminal resolution site, thereby forcing the replication of dimeric self-complementary DNA genomes. Self-complementary AAV hSGSH viral vectors were produced and packaged in AAV serotype 9 capsids. The viral vectors were tested for expression of hSGSH protein and reduction of GAG storage in human MPS IIIA fibroblasts.
  • Self-complementary AAV hSGSH viral vectors were tested in an MPS IIIA mouse model having a missense mutation in the SGSH gene [Bhaumik et al., Glycobiology, 9(12):1389-1396 (1999)] as described in the examples below.
  • EXAMPLE 9
  • MPS IIIA mice were injected at 10 weeks of age with 5×1012 vgp/kg) of scAAV-U1a-hSGSH vector encapsidated in either AAV9 or AAVrh74 serotype. At 10 days post-injection, the mice were euthanized and assays were performed to determine the effects of the treatment.
  • hSGSH transgene expression was assayed. Tissues analyzed included kidney (Kid), heart (Hrt), intestine (Int), skeletal muscle (Mus), lung, brain, Liver (Liv), spleen (Spl) and serum. FIG. 6 shows enzyme expression relative to untreated MPS IIIA mice at the same age (−/−). The scAAV-SGSH vectors reached the CNS and expressed the transgene within days of administration. FIG. 7 shows GAG content measured in the kidney (Kid), heart (Hrt), muscle (Mus), lung, brain, Liver (Liv) and spleen (Spl).
  • Sections of CNS and somatic tissues were stained with the lysosomal marker, Lamp1, revealing clearance of lysosomal storage pathology. Histopathology additionally revealed numerous clear vacuoles present in untreated mice but corrected in treated animals.
  • EXAMPLE 10
  • The therapeutic effects of scAAV-SGSH treatment at a low vector dose were examined.
  • Vector was administered by tail vein injection in MPS IIIA mice at one month of age at an approximately 25-fold lower dose than in Example 9. MPS IIIA mice were treated with 1.7×1011 vgp/kg scAAV9-U1a-hSGSH or 2.7×1011 vgp/kg scAAVrh74-U1a-hSGSH vector.
  • At three months post-injection, expression of SGSH in the CNS was observed by immunofluroescence staining. Correction of astrocytocis, a hallmark of neuroinflamation associated with MPS IIIA pathology, was also observed.
  • At 7-7.5 months age, the animals were tested for learning ability in the Morris water maze. As shown in FIG. 8, compared to untreated controls, treated animals were similar to wt mice in their latency to locate the hidden platform (upper charts) and spent more time in the zone (4) where the platform had been in the previous tests when the platform was removed (lower charts).
  • EXAMPLE 11
  • Therapeutic effects of scAAV treatment at high dose at late stage of disease were also examined.
  • MPS IIIA mice were treated with a high dose (5×1012 vgp/kg) of scAAV9-U1a-hSGSH vector at 6 months of age, after significant neuropathology had already developed. At 7-7.5 months age, the animals were tested for learning ability in the Morris water maze. At 7.5 months of age, the mice were euthanized and tissues assayed for glycosaminoglycan (GAG) content. Tissues analyzed include liver (Liv), kidney (Kid), heart (Hrt), brain, spleen (Spl), lung, skeletal muscle, and intestine.
  • FIG. 9 shows clearance of accumulated GAGs in different tissues, including CNS. FIG. 10 shows, compared to untreated controls, treated animals were similar to wt mice in their latency to locate the hidden platform (upper charts) and spent more time in the zone (4) where the platform had been in the previous tests when the platform was removed (lower charts).
  • While the present invention has been described in terms of various embodiments and examples, it is understood that variations and improvements will occur to those skilled in the art. Therefore, only such limitations as appear in the claims should be placed on the invention.
  • All documents referred to in this application are hereby incorporated by reference in their entirety.

Claims (21)

1. A method of delivering an α-N-acetylglucosamidinase polynucleotide across the blood-brain-barrier comprising the step of systemically administering a rAAV9 comprising a single-stranded genome including the polynucleotide to a patient.
2. A method of delivering an α-N-acetylglucosamidinase polynucleotide to the central nervous system comprising the step of systemically administering a rAAV9 comprising a single-stranded genome including the polynucleotide to a patient.
3.-7. (canceled)
8. A method of delivering an α-N-acetylglucosamidinase polynucleotide to the peripheral nervous system comprising the step of administering a rAAV9 comprising a single-stranded genome including the polynucleotide to a patient.
9.-10. (canceled)
11. A method of treating mucopolysaccharidosis IIIB comprising the step of systemically administering a rAAV9 comprising a single-stranded genome including an α-N-acetylglucosamidinase polynucleotide to a patient.
12. The method of any one of claim 1, 2, 8 or 11 wherein mannitol is administered prior to the administration of the rAAV.
13. (canceled)
14. A rAAV9 comprising a genome encoding α-N-acetylglucosamidinase.
15. (canceled)
16. A composition comprising the rAAV9 of claim 14.
17. A method of delivering an N-sulphoglucosamine sulphohydrolase polynucleotide across the blood-brain-barrier comprising the step of systemically administering a rAAV9 or rh74 comprising a self-complementary genome including the polynucleotide to a patient.
18. A method of delivering an N-sulphoglucosamine sulphohydrolase polynucleotide to the central nervous system comprising the step of systemically administering a rAAV9 or rh74 comprising a self-complementary genome including the polynucleotide to a patient.
19.-23. (canceled)
24. A method of delivering an N-sulphoglucosamine sulphohydrolase polynucleotide to the peripheral nervous system comprising the step of administering a rAAV9 or rh74 comprising a self-complementary genome including the polynucleotide to a patient.
25.-26. (canceled)
27. A method of treating mucopolysaccharidosis IIIA comprising the step of systemically administering a rAAV9 or rh74 comprising a self-complementary genome including an N-sulphoglucosamine sulphohydrolase polynucleotide to a patient.
28. The method of any one of claim 17, 18, 24 or 27 wherein mannitol is administered prior to the administration of the rAAV.
29. (canceled)
30. A rAAV9 or rh74 comprising a genome encoding N-sulphoglucosamine sulphohydrolase.
31. A composition comprising the rAAV9 or rh74 of claim 30.
US13/491,326 2011-06-08 2012-06-07 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders Abandoned US20130039888A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US13/491,326 US20130039888A1 (en) 2011-06-08 2012-06-07 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US13/922,915 US20130323207A1 (en) 2011-06-08 2013-06-20 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US14/950,387 US20160175406A1 (en) 2011-06-08 2015-11-24 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US15/903,861 US20180193431A1 (en) 2011-06-08 2018-02-23 Products and Methods for Delivery of Polynucleotides by Adeno-Associated Virus for Lysosomal Storage Disorders
US16/293,196 US11590210B2 (en) 2011-06-08 2019-03-05 Methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161494635P 2011-06-08 2011-06-08
US13/491,326 US20130039888A1 (en) 2011-06-08 2012-06-07 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/922,915 Division US20130323207A1 (en) 2011-06-08 2013-06-20 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US16/293,196 Continuation US11590210B2 (en) 2011-06-08 2019-03-05 Methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders

Publications (1)

Publication Number Publication Date
US20130039888A1 true US20130039888A1 (en) 2013-02-14

Family

ID=47677669

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/491,326 Abandoned US20130039888A1 (en) 2011-06-08 2012-06-07 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US13/922,915 Abandoned US20130323207A1 (en) 2011-06-08 2013-06-20 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US14/950,387 Abandoned US20160175406A1 (en) 2011-06-08 2015-11-24 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US15/903,861 Pending US20180193431A1 (en) 2011-06-08 2018-02-23 Products and Methods for Delivery of Polynucleotides by Adeno-Associated Virus for Lysosomal Storage Disorders
US16/293,196 Active US11590210B2 (en) 2011-06-08 2019-03-05 Methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders

Family Applications After (4)

Application Number Title Priority Date Filing Date
US13/922,915 Abandoned US20130323207A1 (en) 2011-06-08 2013-06-20 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US14/950,387 Abandoned US20160175406A1 (en) 2011-06-08 2015-11-24 Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US15/903,861 Pending US20180193431A1 (en) 2011-06-08 2018-02-23 Products and Methods for Delivery of Polynucleotides by Adeno-Associated Virus for Lysosomal Storage Disorders
US16/293,196 Active US11590210B2 (en) 2011-06-08 2019-03-05 Methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders

Country Status (1)

Country Link
US (5) US20130039888A1 (en)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015173308A1 (en) * 2014-05-14 2015-11-19 Laboratorios Del Dr. Esteve S.A. Adenoassociated virus vectors for the treatment of lysosomal storage disorders
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
US20160208006A1 (en) * 2015-01-14 2016-07-21 Armagen Technologies, Inc. Methods and compositions for increasing n-acetylglucosaminidase activity in the cns
WO2016126993A1 (en) 2015-02-04 2016-08-11 Washington University Anti-tau constructs
WO2017005806A1 (en) * 2015-07-07 2017-01-12 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
US9975955B2 (en) 2011-12-02 2018-05-22 Armagen, Inc. Methods and compositions for increasing arylsulfatase A activity in the CNS
JP2018515615A (en) * 2015-05-15 2018-06-14 アール. スコット マカイバー Adeno-associated for therapeutic delivery to the central nervous system
US10011651B2 (en) 2009-10-09 2018-07-03 Armagen, Inc. Methods and compositions for increasing iduronate 2-sulfatase activity in the CNS
US10017832B2 (en) 2015-08-25 2018-07-10 Washington University Compositions and methods for site specific recombination at asymmetric sites
US10144783B2 (en) 2006-08-18 2018-12-04 Armagen, Inc. Macromolecular compositions that cross the blood-brain barrier and methods of use thereof
US10202467B2 (en) 2007-07-27 2019-02-12 Armagen, Inc. Methods and compositions for increasing α-L-iduronidase activity in the CNS
WO2019108856A1 (en) * 2017-11-30 2019-06-06 The Trustees Of The University Of Pennsylvania Gene therapy for mucopolysaccharidosis iiib
WO2019108857A1 (en) * 2017-11-30 2019-06-06 The Trustees Of The University Of Pennsylvania Gene therapy for mucopolysaccharidosis iiia
CN110606874A (en) * 2013-07-22 2019-12-24 费城儿童医院 Variant AAV and compositions, methods and uses for gene transfer into cells, organs and tissues
US10876134B2 (en) 2014-12-16 2020-12-29 Board Of Regents Of The University Of Nebraska Gene therapy for juvenile batten disease
US11066456B2 (en) 2016-02-25 2021-07-20 Washington University Compositions comprising TREM2 and methods of use thereof
US11253612B2 (en) 2016-04-15 2022-02-22 The Trustees Of The University Of Pennsylvania Gene therapy for treating mucopolysaccharidosis type II
US11590210B2 (en) 2011-06-08 2023-02-28 Nationwide Children's Hospital, Inc. Methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US11819539B2 (en) 2017-09-22 2023-11-21 The Trustees Of The University Of Pennsylvania Gene therapy for treating Mucopolysaccharidosis type II
US11938193B2 (en) 2016-01-08 2024-03-26 Washington University Compositions comprising chemerin and methods of use thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3169548A1 (en) * 2020-03-23 2021-09-30 Haiyan Fu Aav-naglu vectors for treatment of mucopolysaccharidosis iiib

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070036760A1 (en) * 2003-09-30 2007-02-15 The Trutees Of The University Of Pennsylvania Adeno-associated virus (aav) clades, sequences, vectors containing same, and uses therefor
US20120177605A1 (en) * 2008-12-19 2012-07-12 Nationwide Children's Hospital Inc. Delivery of Polynucleotides Across the Blood-Brain-Barrier Using Recombinant AAV9

Family Cites Families (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
PT728214E (en) 1993-11-09 2004-11-30 Ohio Med College CELL LINES ARE ABLE TO EXPRESS THE ADDITIONAL-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
WO1996017947A1 (en) 1994-12-06 1996-06-13 Targeted Genetics Corporation Packaging cell lines for generation of high titers of recombinant aav vectors
US5863782A (en) * 1995-04-19 1999-01-26 Women's And Children's Hospital Synthetic mammalian sulphamidase and genetic sequences encoding same
FR2737730B1 (en) 1995-08-10 1997-09-05 Pasteur Merieux Serums Vacc PROCESS FOR PURIFYING VIRUSES BY CHROMATOGRAPHY
CA2625279A1 (en) 1995-08-30 1997-03-06 Genzyme Corporation Chromatographic purification of adenovirus and aav
AU6960396A (en) 1995-08-31 1997-03-19 General Hospital Corporation, The Batten disease gene
EP1983057A3 (en) 1995-09-08 2009-01-07 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
KR20000068501A (en) 1996-09-06 2000-11-25 트러스티스 오브 더 유니버시티 오브 펜실바니아 Method for recombinant adeno-associated virus-directed gene therapy
US20040076613A1 (en) 2000-11-03 2004-04-22 Nicholas Mazarakis Vector system
EP1009808B1 (en) 1997-09-05 2012-12-05 Genzyme Corporation Methods for generating high titer helper-free preparations of recombinant aav vectors
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
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
WO2001036603A2 (en) 1999-11-17 2001-05-25 Avigen, Inc. Recombinant adeno-associated virus virions for the treatment of lysosomal disorders
WO2001083692A2 (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
US20030083299A1 (en) 2000-11-04 2003-05-01 Ferguson Ian A. Non-invasive delivery of polypeptides through the blood-brain barrier
US6841357B1 (en) 2001-03-30 2005-01-11 Stratagene California Method and kits for titering adeno-associated virus vectors
US20020192688A1 (en) 2001-04-05 2002-12-19 Xiaoming Yang Imaging nucleic acid delivery
EP2573170B1 (en) 2001-12-17 2017-12-20 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) serotype 9 sequences, vectors containing same, and uses therefor
US6998118B2 (en) 2001-12-21 2006-02-14 The Salk Institute For Biological Studies Targeted retrograde gene delivery for neuronal protection
WO2004098648A1 (en) 2003-05-01 2004-11-18 Genzyme Corporation Gene therapy for neurometabolic disorders
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
WO2005056807A2 (en) 2003-12-04 2005-06-23 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services, National Institutes Of Health Bovine adeno-associated viral (baav) vector and uses thereof
JP5003480B2 (en) 2004-03-06 2012-08-15 ブロードベクター リミテッド Enzyme-prodrug treatment for prosthetic joint repair
US8679460B2 (en) 2004-03-11 2014-03-25 Mallinckrodt Llc Low osmolar X-ray contrast media formulations
EP2359865B1 (en) 2005-04-07 2013-10-02 The Trustees of The University of Pennsylvania Method of increasing the function of an AAV vector
CA2614754A1 (en) 2005-07-12 2007-01-18 Renovo Ltd Pharmaceutical compositions comprising a tgf-beta superfamily member
US7867484B2 (en) 2006-01-27 2011-01-11 University Of North Carolina At Chapel Hill Heparin and heparan sulfate binding chimeric vectors
US20070280906A1 (en) 2006-06-03 2007-12-06 Ognjen Petras Method to generate mirrored adenoassociated viral vectors
EP3023500B1 (en) 2006-06-21 2020-02-12 uniQure IP B.V. Insect cells for the production of aav vectors
EP3252161B1 (en) 2007-06-06 2021-11-17 Genzyme Corporation Gene therapy for lysosomal storage diseases
US8299215B2 (en) 2007-07-14 2012-10-30 University Of Iowa Research Foundation Methods and compositions for treating brain diseases
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
WO2009137006A2 (en) 2008-04-30 2009-11-12 The University Of North Carolina At Chapel Hill Directed evolution and in vivo panning of virus vectors
US9415121B2 (en) 2008-12-19 2016-08-16 Nationwide Children's Hospital Delivery of MECP2 polynucleotide using recombinant AAV9
US11219696B2 (en) 2008-12-19 2022-01-11 Nationwide Children's Hospital Delivery of polynucleotides using recombinant AAV9
EP3421603B1 (en) 2009-05-02 2021-10-06 Genzyme Corporation Gene therapy for neurodegenerative disorders
WO2011112902A2 (en) 2010-03-12 2011-09-15 The Board Of Trustees Of The Leland Stanford Junior University Nonviral gene delivery vector iopamidol, protamine, ethiodized oil reagent (viper)
TWI688395B (en) 2010-03-23 2020-03-21 英翠克頌公司 Vectors conditionally expressing therapeutic proteins, host cells comprising the vectors, and uses thereof
WO2011133890A1 (en) 2010-04-23 2011-10-27 University Of Massachusetts Cns targeting aav vectors and methods of use thereof
EP2394667A1 (en) * 2010-06-10 2011-12-14 Laboratorios Del Dr. Esteve, S.A. Vectors and sequences for the treatment of diseases
CN103189507A (en) 2010-10-27 2013-07-03 学校法人自治医科大学 Adeno-associated virus virions for transferring genes into neural cells
AU2010366066B2 (en) * 2010-12-22 2016-01-14 Fondazione Telethon Therapeutic strategies to treat CNS pathology in mucopolysaccharidoses
US20130039888A1 (en) 2011-06-08 2013-02-14 Nationwide Children's Hospital Inc. Products and methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
AU2012347765B2 (en) 2011-12-06 2017-05-04 Research Institute At Nationwide Childrens's Hospital Non-ionic, low osmolar contrast agents for delivery of antisense oligonucleotides and treatment of disease
ES2684222T3 (en) 2012-08-01 2018-10-01 Nationwide Children's Hospital Intrathecal administration of recombinant adeno-associated 9 virus
JP7384797B2 (en) 2017-11-30 2023-11-21 ザ・トラステイーズ・オブ・ザ・ユニバーシテイ・オブ・ペンシルベニア Gene therapy for mucopolysaccharidosis type IIIB
MX2020005663A (en) 2017-11-30 2020-08-20 Univ Pennsylvania Gene therapy for mucopolysaccharidosis iiia.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070036760A1 (en) * 2003-09-30 2007-02-15 The Trutees Of The University Of Pennsylvania Adeno-associated virus (aav) clades, sequences, vectors containing same, and uses therefor
US20120177605A1 (en) * 2008-12-19 2012-07-12 Nationwide Children's Hospital Inc. Delivery of Polynucleotides Across the Blood-Brain-Barrier Using Recombinant AAV9

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Cressant et al The Journal of Neuroscience, November 10, 2004, 24(45):10229 -10239 *
Duque Mol. Ther. 17:1187-1196 (2009) *
Fu et al Molecular Genetics and Metabolism, (February 2011) Vol. 102, No. 2, pp. S18-S19. Abstract Number: 118.Meeting Info: 7th Annual Research Meeting of the Lysosomal Disease Network, WORLD Symposium 2011. Las Vegas, NV (abstract only), page 1. , *
Fu et al Molecular Therapy, (May 2011) Vol. 19, Supp. SUPPL. 1, pp. S131. Abstract Number: 337. Meeting Info: 14th Annual Meeting of the American Society of Gene and Cell Therapy. Seattle, WA, United States. 18 May 2011-21 May 2011, abstract only page 1. *
Fu et al Molecular Therapy, 2011, 19(6), 1025-1033, 2011, published online, 3/8/2011 *
Manfredsson et al Molecular Therapy, 2009, 17, 403-405 *
McCarty et al (Gene Therapy (2009) 16, 1340-1352, *
Ruzo et al XVlll Annual Congress of the European Society of Gene and Cell Therapy: 1389, abstract Or 96, October 22-25, 2010 *

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11155631B2 (en) 2006-08-18 2021-10-26 Armagen, Inc. Macromolecular compositions that cross the blood-brain barrier and methods of use thereof
US10144783B2 (en) 2006-08-18 2018-12-04 Armagen, Inc. Macromolecular compositions that cross the blood-brain barrier and methods of use thereof
US11512145B2 (en) 2007-07-27 2022-11-29 Armagen, Inc. Methods and compositions for increasing alpha-L-iduronidase activity in the CNS
US10202467B2 (en) 2007-07-27 2019-02-12 Armagen, Inc. Methods and compositions for increasing α-L-iduronidase activity in the CNS
US10011651B2 (en) 2009-10-09 2018-07-03 Armagen, Inc. Methods and compositions for increasing iduronate 2-sulfatase activity in the CNS
US11028156B2 (en) 2009-10-09 2021-06-08 Armagen, Inc. Methods and compositions for increasing iduronate 2-sulfatase activity in the CNS
US11590210B2 (en) 2011-06-08 2023-02-28 Nationwide Children's Hospital, Inc. Methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US9975955B2 (en) 2011-12-02 2018-05-22 Armagen, Inc. Methods and compositions for increasing arylsulfatase A activity in the CNS
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
US20180036388A1 (en) * 2013-05-15 2018-02-08 Regenxbio Inc. Adeno-associated virus mediated gene transfer to the central nervous system
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
CN110606874A (en) * 2013-07-22 2019-12-24 费城儿童医院 Variant AAV and compositions, methods and uses for gene transfer into cells, organs and tissues
KR20170026358A (en) * 2014-05-14 2017-03-08 라보라토리오스 델 드라. 에스테브.에스.에이. Adenoassociated virus vectors for the treatment of lysosomal storage disorders
CN107002095A (en) * 2014-05-14 2017-08-01 埃斯蒂维实验室股份有限公司 Adeno-associated virus vector for treating lysosomal storage disease
KR102446169B1 (en) * 2014-05-14 2022-09-22 에스테베 파마슈티칼스 에스에이 Adenoassociated virus vectors for the treatment of lysosomal storage disorders
AU2015261519B2 (en) * 2014-05-14 2021-08-05 Esteve Pharmaceuticals, S.A. Adenoassociated virus vectors for the treatment of lysosomal storage disorders
JP2017523802A (en) * 2014-05-14 2017-08-24 ラボラトリオス デル ドクター エステヴェ ソシエダッド アノニマ Adeno-associated virus vector for treatment of lysosomal storage diseases
WO2015173308A1 (en) * 2014-05-14 2015-11-19 Laboratorios Del Dr. Esteve S.A. Adenoassociated virus vectors for the treatment of lysosomal storage disorders
US11149285B2 (en) 2014-05-14 2021-10-19 Universitat Autónoma De Barcelona Adenoassociated virus vectors for the treatment of lysosomal storage disorders
US10876134B2 (en) 2014-12-16 2020-12-29 Board Of Regents Of The University Of Nebraska Gene therapy for juvenile batten disease
US20160208006A1 (en) * 2015-01-14 2016-07-21 Armagen Technologies, Inc. Methods and compositions for increasing n-acetylglucosaminidase activity in the cns
US10538589B2 (en) * 2015-01-14 2020-01-21 Armagen Inc. Methods and compositions for increasing N-acetylglucosaminidase (NAGLU) activity in the CNS using a fusion antibody comprising an anti-human insulin receptor antibody and NAGLU
EP4059961A2 (en) 2015-02-04 2022-09-21 Washington University Anti-tau constructs
WO2016126993A1 (en) 2015-02-04 2016-08-11 Washington University Anti-tau constructs
EP4000631A1 (en) * 2015-05-15 2022-05-25 REGENXBIO Inc. Adeno-associated for therapeutic delivery to central nervous system
JP2018515615A (en) * 2015-05-15 2018-06-14 アール. スコット マカイバー Adeno-associated for therapeutic delivery to the central nervous system
EP3294323A4 (en) * 2015-05-15 2019-01-16 McIvor, R. Scott Adeno-associated for therapeutic delivery to central nervous system
AU2016263119B2 (en) * 2015-05-15 2021-08-12 Regents Of The University Of Minnesota Adeno-associated for therapeutic delivery to central nervous system
JP2021167332A (en) * 2015-05-15 2021-10-21 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ Adeno-associated for therapeutic delivery to central nervous system
WO2017005806A1 (en) * 2015-07-07 2017-01-12 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
US10801040B2 (en) 2015-07-07 2020-10-13 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
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
US10017832B2 (en) 2015-08-25 2018-07-10 Washington University Compositions and methods for site specific recombination at asymmetric sites
US11938193B2 (en) 2016-01-08 2024-03-26 Washington University Compositions comprising chemerin and methods of use thereof
US11066456B2 (en) 2016-02-25 2021-07-20 Washington University Compositions comprising TREM2 and methods of use thereof
US11253612B2 (en) 2016-04-15 2022-02-22 The Trustees Of The University Of Pennsylvania Gene therapy for treating mucopolysaccharidosis type II
US11819539B2 (en) 2017-09-22 2023-11-21 The Trustees Of The University Of Pennsylvania Gene therapy for treating Mucopolysaccharidosis type II
WO2019108857A1 (en) * 2017-11-30 2019-06-06 The Trustees Of The University Of Pennsylvania Gene therapy for mucopolysaccharidosis iiia
WO2019108856A1 (en) * 2017-11-30 2019-06-06 The Trustees Of The University Of Pennsylvania Gene therapy for mucopolysaccharidosis iiib
US11555206B2 (en) 2017-11-30 2023-01-17 The Trustees Of The University Of Pennsylvania Gene therapy for mucopolysaccharidosis IIIA
US11723989B2 (en) 2017-11-30 2023-08-15 The Trustees Of The University Of Pennsylvania Gene therapy for mucopolysaccharidosis IIIB

Also Published As

Publication number Publication date
US20180193431A1 (en) 2018-07-12
US20160175406A1 (en) 2016-06-23
US20130323207A1 (en) 2013-12-05
US20200000887A1 (en) 2020-01-02
US11590210B2 (en) 2023-02-28

Similar Documents

Publication Publication Date Title
US11590210B2 (en) Methods for delivery of polynucleotides by adeno-associated virus for lysosomal storage disorders
US20200297872A1 (en) Delivery of polynucleotides using recombinant aav9
US11311634B2 (en) Intrathecal delivery of recombinant Adeno-associated virus 9
US20220125952A1 (en) Delivery of polynucleotides using recombinant aav9
US20120177605A1 (en) Delivery of Polynucleotides Across the Blood-Brain-Barrier Using Recombinant AAV9
US20220267768A1 (en) Recombinant virus products and methods for inducing dux4 exon skipping
US20230227515A1 (en) Optimized gene therapy for targeting muscle in muscle diseases
US20240115735A1 (en) Materials and methods for the treatment of lysosomal acid lipase deficiency (lal-d)
US20230149564A1 (en) Aav-naglu vectors for treatment of mucopolysaccharidosis iiib
WO2024011115A1 (en) Adeno-associated virus delivery of cln1 polynucleotide

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONWIDE CHILDREN'S HOSPITAL, INC., OHIO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MCCARTY, DOUGLAS M.;FU, HAIYAN;REEL/FRAME:030186/0059

Effective date: 20110720

STCB Information on status: application discontinuation

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