US20210085759A1 - Methods to treat pompe disease using a recombinant adeno-associated virus - Google Patents

Methods to treat pompe disease using a recombinant adeno-associated virus Download PDF

Info

Publication number
US20210085759A1
US20210085759A1 US17/118,353 US202017118353A US2021085759A1 US 20210085759 A1 US20210085759 A1 US 20210085759A1 US 202017118353 A US202017118353 A US 202017118353A US 2021085759 A1 US2021085759 A1 US 2021085759A1
Authority
US
United States
Prior art keywords
administered
vector
aav
raav
human
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
US17/118,353
Inventor
R. Scott McIvor
Lalitha R. Belur
Walter Low
Carolyn Fairbanks
Karen Kozarsky
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Minnesota
Regenxbio Inc
Original Assignee
University of Minnesota
Regenxbio 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 University of Minnesota, Regenxbio Inc filed Critical University of Minnesota
Priority to US17/118,353 priority Critical patent/US20210085759A1/en
Assigned to REGENTS OF THE UNIVERSITY OF MINNESOTA reassignment REGENTS OF THE UNIVERSITY OF MINNESOTA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LOW, WALTER, BELUR, Lalitha R., MCIVOR, R. SCOTT, FAIRBANKS, Carolyn
Assigned to REGENXBIO INC. reassignment REGENXBIO INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KOZARSKY, KAREN
Publication of US20210085759A1 publication Critical patent/US20210085759A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • 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)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0085Brain, e.g. brain implants; Spinal cord
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • C12N15/864Parvoviral vectors, e.g. parvovirus, densovirus
    • C12N15/8645Adeno-associated virus
    • 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
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01076L-Iduronidase (3.2.1.76)
    • HELECTRICITY
    • H05ELECTRIC TECHNIQUES NOT OTHERWISE PROVIDED FOR
    • H05KPRINTED CIRCUITS; CASINGS OR CONSTRUCTIONAL DETAILS OF ELECTRIC APPARATUS; MANUFACTURE OF ASSEMBLAGES OF ELECTRICAL COMPONENTS
    • H05K999/00PRINTED CIRCUITS; CASINGS OR CONSTRUCTIONAL DETAILS OF ELECTRIC APPARATUS; MANUFACTURE OF ASSEMBLAGES OF ELECTRICAL COMPONENTS dummy group
    • H05K999/99PRINTED CIRCUITS; CASINGS OR CONSTRUCTIONAL DETAILS OF ELECTRIC APPARATUS; MANUFACTURE OF ASSEMBLAGES OF ELECTRICAL COMPONENTS dummy group dummy group
    • 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
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/007Vector systems having a special element relevant for transcription cell cycle specific enhancer/promoter combination

Definitions

  • MPSs mucopolysaccharidoses
  • GAG glycosaminoglycan
  • Manifestations of varying severity include organomegaly, skeletal dysplasias, cardiac and pulmonary obstruction and neurological deterioration.
  • MPS I deficiency of iduronidase (IDUA), severity ranges from mild (Scheie syndrome) to moderate (Hurler-Scheie) to severe (Hurler syndrome), with the latter resulting in neurologic deficiency and death by age 15 (Muenzer, 2004; Munoz-Rojas et al., 2008).
  • Therapies for MPSs have been for the most part palliative.
  • Hurler syndrome for which allogeneic hematopoietic stem cell transplantation (HSCT) has exhibited efficacy (Krivit, 2004; Orchard et al., 2007; Peters et al., 2003).
  • ERT enzyme replacement therapy
  • HSCT and ERT result in the clearing of storage materials and improved peripheral conditions, although some problems persist after treatment (skeletal, cardiac, corneal clouding).
  • the primary challenge in these cellular and enzyme therapies is effectiveness in addressing neurological manifestations, as peripherally administered enzyme does not penetrate the blood-brain barrier and HSCT has been found to be of benefit for some, but not all, MPS's.
  • MPS I has been one of the most extensively studied of the MPS diseases for development of cellular and molecular therapies.
  • the effectiveness of allogeneic HSCT is most likely the result of metabolic cross-correction, whereby the missing enzyme is released from donor-derived cells and subsequently taken up by host cells and trafficked to lysosomes, where the enzyme contributes to lysosomal metabolism (Fratantoni et al., 1968). Clearing of GAG storage materials is subsequently observed in peripheral organs such as liver and spleen, there is relief from cardiopulmonary obstruction and improvement in corneal clouding (Orchard et al., 2007). Of particular importance is the effect of allogeneic stem cell transplantation on the emergence of neurologic manifestations in the MPS diseases.
  • the level of enzyme provided to CNS tissues is thus limited to that amount expressed and released from donor-derived cells engrafting in the brain. While such engraftment is of great benefit for MPS I, recipients nonetheless continue to exhibit below normal IQ and impaired neurocognitive capability (Ziegler and Shapiro, 2007).
  • Methods of preventing, inhibiting, and/or treating one or more symptoms associated with a disease of the central nervous system (CNS) in a mammal in need thereof are described.
  • the methods involve delivering to the CNS of a mammal in need of treatment a composition comprising an effective amount of a recombinant adeno-associated virus (rAAV) vector comprising an open reading frame encoding a gene product, e.g., a therapeutic gene product.
  • rAAV recombinant adeno-associated virus
  • Target gene products that may be encoded by an rAAV vector include, but are not limited to, alpha-L-iduronidase, iduronate-2-sulfatase, heparan sulfate sulfatase, N-acetyl-alpha-D-glucosaminidase, beta-hexosaminidase, alpha-galactosidase, betagalactosidase, beta-glucuronidase or glucocerebrosidase.
  • Diseases that may be prevented, inhibited or treated using the methods disclosed herein include, but are not limited to, mucopolysaccharidosis type I disorder, a mucopolysaccharidosis type II disorder, or a mucopolysaccharidosis type VII disorder.
  • the AAV vector can be administered in a variety of ways to ensure that it is delivered to the CNS/brain, and that the transgene is successfully transduced in the subject's CNS/brain. Routes of delivery to the CNS/brain include, but are not limited to intrathecal administration, intracranial administration, e.g., intracerebroventricular administration, or lateral cerebro ventricular administration), intranasal administration, endovascular administration, and intraparenchymal administration.
  • the methods involve delivering to the CNS of an adult mammal in need of treatment a composition comprising an effective amount of a rAAV-9 vector comprising an open reading frame encoding a gene. In one embodiment, the methods involve delivering to the CNS of an adult mammal in need of treatment a composition comprising an effective amount of a rAAV-9 vector comprising an open reading frame encoding IDUA.
  • the working examples demonstrate that co-therapy to induce immunosuppression or immunotolerization, or treatment of immunodeficient animals, can achieve even higher levels of IDUA enzyme expression and activity.
  • patients with genotypes that promote an immune response that neutralizes enzyme activity are treated with an immunosuppressant in addition to the rAAV vector comprising an open reading frame encoding a gene product, such as IDUA.
  • Neonatal IDUA ⁇ / ⁇ mice are not immunocompetent.
  • administration of IDUA expressing AAV-8 to neonatal IDUA ⁇ / ⁇ mice resulted in IDUA expression (Wolf et al., 2011), thus tolerizing the animals to IDUA.
  • IDUA expression Wang et al., 2011
  • AAV-IDUA serotype 9 was administered by direct injection into the lateral ventricles of adult IDUA-deficient mice that were either immunocompetent, immunodeficient (NODSCID/IDUA ⁇ / ⁇ ), immunosuppressed with cyclophosphamide (CP), or immunotolerized by weekly injection of human iduronidase protein (Aldurazyme) starting at birth.
  • CP immunosuppressed animals were also administered AAV9-IDUA by intranasal infusion, by intrathecal injection, and by endovascular infusion with and without mannitol to disrupt the blood-brain barrier.
  • mice Animals were sacrificed at 8 weeks after vector administration, and brains were harvested and microdissected for evaluation of IDUA enzymatic activity, tissue glycosaminoglycans, and IDUA vector sequences in comparison with normal and affected control mice. Results from these studies show that numerous routes for AAV vector administration directly to the CNS may be employed, e.g., so as to achieve higher levels of protein delivery and/or enzyme activity in the CNS.
  • routes for AAV vector administration directly to the CNS may be employed, e.g., so as to achieve higher levels of protein delivery and/or enzyme activity in the CNS.
  • the brain is an immunopriviledged site, administration of an immunosuppressant or immunotolerization may increase the activity found in the brain after AAV administration. Higher levels of expression per administration and/or less invasive routes of administration are clinically more palatable to patients.
  • the invention includes the use of recombinant AAV (rAAV) vectors that encode a gene product with therapeutic effects when expressed in the CNS of a mammal.
  • the mammal is an immunocompetent mammal with a disease or disorder of the CNS (a neurologic disease).
  • An “immunocompetent” mammal as used herein is a mammal of an age where both cellular and humoral immune responses are elicited after exposure to an antigenic stimulus, by upregulation of Th1 functions or IFN- ⁇ production in response to polyclonal stimuli, in contrast to a neonate which has innate immunity and immunity derived from the mother, e.g., during gestation or via lactation.
  • an adult mammal that does not have an immunodeficiency disease is an example of an immunocompetent mammal.
  • an immunocompetent human is typically at least 1, 2, 3, 4, 5 or 6 months of age, and includes adult humans without an immunodeficiency disease.
  • the AAV is administered intrathecally.
  • the MV is administered intracranially (e.g., intracerebroventricularly).
  • the AAV is administered intranasally, with or without a permeation enhancer.
  • the AAV is administered endovascularly, e.g., carotid artery administration, with or without a permeation enhancer.
  • the mammal that is administered the AAV is immunodeficient or is subjected to immunotolerization or immune suppression, e.g., to induce higher levels of therapeutic protein expression relative to a corresponding mammal that is administered the MV but not subjected to immunotolerization or immune suppression.
  • an immune suppressive agent is administered to induce immune suppression.
  • the mammal that is administered the AAV is not subjected to immunotolerization or immune suppression (e.g., administration of the AAV alone provides for the therapeutic effect).
  • the invention provides a method to prevent, inhibit or treat one or more symptoms associated with a disease or disorder of the central nervous system in a mammal in need thereof.
  • the method includes intrathecally, e.g., to the lumbar region, or intracerebroventricularly, e.g., to the lateral ventricle, administering to the mammal a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms.
  • the gene product is a lysosomal storage enzyme.
  • the mammal is an immunocompetent adult.
  • the rAAV vector is an AAV-1, MV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, MV rh10, or AAV-9 vector.
  • the mammal is a human.
  • multiple doses are administered.
  • the composition is administered weekly, monthly or two or more months apart.
  • the method includes intrathecally, e.g., to the lumbar region, administering to a mammal a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms, and optionally administering a permeation enhancer.
  • the permeation enhancer is administered before the composition.
  • the composition comprises a permeation enhancer.
  • the permeation enhancer is administered after the composition.
  • the gene product is a lysosomal storage enzyme.
  • the mammal is an immunocompetent adult.
  • the rAAV vector is an AAV-1, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV rh10, or AAV-9 vector.
  • the mammal is a human.
  • multiple doses are administered.
  • the composition is administered weekly, monthly or two or more months apart.
  • the mammal that is intrathecally administered the AAV is not subjected to immunotolerization or immune suppression (e.g., administration of the AAV alone provides for the therapeutic effect).
  • the mammal that is intrathecally administered the AAV is immunodeficient or is subjected to immunotolerization or immune suppression, e.g., to induce higher levels of therapeutic protein expression relative to a corresponding mammal that is intrathecally administered the AAV but not subjected to immunotolerization or immune suppression.
  • the method includes intracerebroventricularly, e.g., to the lateral ventricle, administering to an immunocompetent mammal a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms.
  • the gene product is a lysosomal storage enzyme.
  • the rAAV vector is an AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV rh10, or AAV-9 vector. In one embodiment, the rAAV vector is not a rAAV-5 vector.
  • the mammal is a human. In one embodiment, multiple doses are administered. In one embodiment, the composition is administered weekly, monthly or two or more months apart. In one embodiment, the mammal that is intracerebroventricularly administered the AAV is not subjected to immunotolerization or immune suppression (e.g., administration of the AAV alone provides for the therapeutic effect).
  • the mammal that is intracerebroventricularly administered the AAV is immunodeficient or is subjected to immunotolerization or immune suppression, e.g., to induce higher levels of therapeutic protein expression relative to a corresponding mammal that is intracerebroventricularly administered the AAV but not subjected to immunotolerization or immune suppression
  • the mammal is immunotolerized to the gene product before the composition comprising the AAV is administered.
  • the method includes endovascularly administering to the mammal a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms, and an effective amount of a permeation enhancer.
  • the composition comprises the permeation enhancer.
  • the permeation enhancer comprises mannitol, sodium glycocholate, sodium taurocholate, sodium deoxycholate, sodium salicylate, sodium caprylate, sodium caprate, sodium lauryl sulfate, polyoxyethylene-9-laurel ether, or EDTA.
  • the gene product is a lysosomal storage enzyme.
  • the mammal is an immunocompetent adult.
  • the rAAV vector is an AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, MV rh10, or AAV-9 vector. In one embodiment, the rAAV vector is not a rAAV-5 vector.
  • the mammal is a human. In one embodiment, multiple doses are administered. In one embodiment, the composition is administered weekly. In one embodiment, the composition is administered weekly, monthly or two or more months apart. In one embodiment, the mammal that is endovascularly administered the AAV is not subjected to immunotolerization or immune suppression (e.g., administration of the MV provides for the therapeutic effect). In one embodiment, the mammal that is endovascularly administered the AAV is immunodeficient or is subjected to immunotolerization or immune suppression, e.g., to induce higher levels of therapeutic protein expression relative to a corresponding mammal that is endovascularly administered the AAV but not subjected to immunotolerization or immune suppression.
  • the method includes intranasally administering to a mammal a composition comprising an effective amount of a rAAV-9 vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms, and optionally administering a permeation enhancer.
  • intranasal delivery may be accomplished as described in U.S. Pat. No. 8,609,088, the disclosure of which is incorporated by reference herein.
  • the permeation enhancer is administered before the composition.
  • the composition comprises a permeation enhancer.
  • the permeation enhancer is administered after the composition.
  • the gene product is a lysosomal storage enzyme.
  • the mammal is an immunocompetent adult. In one embodiment, the mammal is a human. In one embodiment, multiple doses are administered. In one embodiment, the composition is administered weekly, monthly or two or more months apart. In one embodiment, the mammal that is intranasally administered the AAV is not subjected to immunotolerization or immune suppression. In one embodiment, the mammal that is intranasally administered the AAV is subjected to immunotolerization or immune suppression, e.g., to induce higher levels of IDUA protein expression relative to a corresponding mammal that is intranasally administered the AAV but not subjected to immunotolerization or immune suppression.
  • the method includes administering to the mammal a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms, and an immune suppressant.
  • the immune suppressant comprises cyclophosphamide.
  • the immune suppressant comprises a glucocorticoid, cytostatic agents including an alkylating agent or an anti-metabolite such as methotrexate, azathioprine, mercaptopurine or a cytotoxic antibiotic, an antibody, or an agent active on immunophilin.
  • cytostatic agents including an alkylating agent or an anti-metabolite such as methotrexate, azathioprine, mercaptopurine or a cytotoxic antibiotic, an antibody, or an agent active on immunophilin.
  • the immune suppressant comprises a nitrogen mustard, nitrosourea, a platinum compound, methotrexate, azathioprine, mercaptopurine, fluorouracil, dactinomycin, an anthracyclin, mitomycin C, bleomycin, mithramycin, IL2-receptor- (CD25-) or CD3-directed antibodies, anti-IL-2 antibodies, cyclosporin, tacrolimus, sirolimus, IFN- ⁇ , IFN- ⁇ , an opioid, or TNF- ⁇ (tumor necrosis factor-alpha) binding agents such as infliximab (Remicade), etanercept (Enbrel), or adalimumab (Humira).
  • the rAAV and the immune suppressant are co-administered. In one embodiment, the rAAV is administered before and optionally after the immune suppressant. In one embodiment, the immune suppressant is administered before the rAAV. In one embodiment, the rAAV and the immune suppressant are intrathecally administered. In one embodiment, the rAAV and the immune suppressant are intracerebroventricularly administered. In one embodiment, the rAAV is intrathecally administered and the immune suppressant is intravenously administered. In one embodiment, the gene product is a lysosomal storage enzyme. In one embodiment, the mammal is an adult.
  • the rAAV vector is an AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV rh10, or AAV-9 vector.
  • the mammal is a human.
  • multiple doses are administered.
  • the composition is administered weekly. In one embodiment, the composition is administered weekly, monthly or two or more months apart.
  • the invention also provides a method to prevent, inhibit or treat one or more symptoms associated with a disease of the central nervous system in a mammal in need thereof.
  • a mammal immunotolerized to a gene product that is associated with the disease is administered a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms.
  • the gene product is a lysosomal storage enzyme.
  • the mammal is an adult.
  • the rAAV vector is an AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, MV rh10, or AAV-9 vector.
  • the mammal is a human.
  • multiple doses are administered.
  • the composition is administered weekly.
  • Gene products that may be encoded by rAAV vectors include, but are not limited to, alpha-L-iduronidase, iduronate-2-sulfatase, heparan sulfate sulfatase, N-acetyl-alpha-D-glucosaminidase, beta-hexosaminidase, alpha-galactosidase, betagalactosidase, beta-glucuronidase, glucocerebrosidase, fibroblast growth factor-2 (FGF-2), brain derived growth factor (BDGF), neurturin, glial derived growth factor (GDGF), tyrosine hydroxylase, dopamine decarboxylase, or glutamic acid decarboxylase.
  • FGF-2 fibroblast growth factor-2
  • BDGF brain derived growth factor
  • GDGF neurturin
  • GDGF glial derived growth factor
  • tyrosine hydroxylase do
  • Diseases that have one or more neurologic symptoms that may be prevented, inhibited or treated using the methods disclosed herein include, but are not limited to, Adrenoleukodystrophy, Alzheimer disease, Amyotrophic lateral sclerosis, Angelman syndrome, Ataxia telangiectasia, Charcot-Marie-Tooth syndrome, Cockayne syndrome, Deafness, Duchenne muscular dystrophy, Epilepsy, Essential tremor, Fragile X syndrome, Friedreich's ataxia, Gaucher disease, Huntington disease, Lesch-Nyhan syndrome, Maple syrup urine disease, Menkes syndrome, Myotonic dystrophy, Narcolepsy, Neurofibromatosis, Niemann-Pick disease, Parkinson disease, Phenylketonuria, Prader-Willi syndrome, Refsum disease, Rett syndrome, Spinal muscular atrophy, Spinocerebellar ataxia, Tangier disease, Tay-Sachs disease, Tuberous sclerosis, Von Hippel-Lindau syndrome, Williams syndrome, Wilson'
  • the disease is a lysosomal storage disease, e.g., a lack or deficiency in a lysosomal storage enzyme.
  • Lysosomal storage diseases include, but are not limited to, mucopolysaccharidosis (MPS) diseases, for instance, mucopolysaccharidosis type I, e.g., Hurler syndrome and the variants Scheie syndrome and Hurler-Scheie syndrome (a deficiency in alpha-L-iduronidase); Hunter syndrome (a deficiency of iduronate-2-sulfatase); mucopolysaccharidosis type III, e.g., Sanfilippo syndrome (A, B, C or D; a deficiency of heparan sulfate sulfatase, N-acetyl-alpha-D-glucosaminidase, acetyl CoA:alpha-glucosaminide N-acetyl transferase
  • the methods described herein involve delivering to the CNS of an immunocompetent human in need of treatment a composition comprising an effective amount of a rAAV-9 vector comprising an open reading frame encoding a IDUA.
  • Routes of administration to the CNS/brain include, but are not limited to intrathecal administration, intracranial administration, e.g., intracerebroventricular administration or lateral cerebro ventricular administration, intranasal administration, endovascular administration, and intraparenchymal administration.
  • viral vectors may be employed in the methods of the invention, e.g., viral vectors such as retrovirus, lentivirus, adenovirus, semliki forest virus or herpes simplex virus vectors.
  • FIG. 1 Experimental design for iduronidase-deficient adult mice administered IDUA-AAV either intracerebroventricularly (ICV) (right lateral ventricle) or intrathecally (lumbar area).
  • IDUA-AAV intracerebroventricularly
  • CP cyclophosphamide
  • aldurazyme human iduonidase protein
  • injections 10 microliters, 3 ⁇ 10 11 particles
  • FIGS. 2A-B IDUA activity in immunodeficient, IDUA deficient animals after ICV injection.
  • FIGS. 3A-B IDUA activity in immunocompetent animals administered AAV vector by ICV route and treated with cyclophosphamide.
  • FIGS. 4A-B IDUA activity in immunocompetent animals administered AAV vector by IT route and treated with cyclophosphamide.
  • FIGS. 5A-B IDUA activity in immunotolerized immunocompetent animals administered AAV vector ICV.
  • FIG. 6 Compilation of all mean levels of IDUA activity for side-by-side comparison.
  • FIGS. 7A-E Data are grouped according the area of the brain. A) hippocampus, B) cerebellum, C) cortex, D) striatum, and E) rest of brain.
  • FIGS. 8A-D Assay for GAG storage material in the different sections of the brain for all four of the test groups.
  • FIG. 9 Schematic of experimental design. All mice were adult MPS1 (IDUA deficient) mice injected with 10 microliters (3 ⁇ 10 11 particles) of AAV.
  • FIGS. 10A-B Intracranial infusion of AAV9IDUA into immunodeficient MPS I mice.
  • Adult animals were injected with 10 11 vector genomes and evaluated for iduronidase expression in the brain after 10 weeks.
  • Enzyme activity levels in the brain were significantly higher than in the brains of wild type animals, and ranged from 30- to 300-fold higher than wild type.
  • FIGS. 11A-B Intracranial administration of AAV9IDUA in immunocompetent, IDUA deficient mice.
  • Adult animals were injected with 10 11 vector genomes, and immunosuppressed by weekly injection of cyclophosphamide (CP).
  • CP injections were terminated at 6 weeks post vector injection due to poor health, and the animals were sacrificed at 8 weeks post-injection. Brains were microdissected and assayed for IDUA enzyme activity.
  • FIGS. 12A-B Intracranial infusion of AAV9IDUA into immunotolerized MPS I mice.
  • MPS 1 mice were tolerized with either a single dose of Aldurazyme at birth or multiple doses administered weekly, starting at birth. Mice were infused with vector at 4 months, and sacrificed at 11 weeks after injection. Brains were microdissected and analyzed for iduronidase expression. Enzyme activities ranged from an average of 10- to 1000-fold higher than wild type levels.
  • FIGS. 13A-B Intrathecal administration of AAV9IDUA in immunocompetent, IDUA deficient animals.
  • Adult MPS I mice were injected with AAV9IDUA intrathecally, followed by a weekly immunosuppressive regimen of cyclophosphamide. Animals were sacrificed at 11 weeks post-injection, and then brains and spinal cords were analyzed for IDUA enzyme activity.
  • FIGS. 14A-B Intrathecal infusion of AAV9IDUA in immunotolerized MPS I mice. IDUA deficient animals were tolerized at birth with a single dose of Aldurazyme or multiple doses administered weekly starting at birth. At 4 months of age animals were infused intrathecally with AAV9IDUA vector, and at 10 weeks post-injection animals were sacrificed, brains microdissected and assayed for iduronidase activity. There was restoration of enzyme activity in all parts of the brain, with activities in the cerebellum ranging from 200- to 1500-fold higher than wild type levels. Levels of enzyme activity in the olfactory bulb and cerebellum (to the right of the dashed line) correspond to the right Y-axis.
  • FIGS. 15A-B Intrathecal infusion of AAV9IDUA in immunocompetent MPS I animals. Control MPS I animals were injected with AAV9IDUA vector, but were not immunosuppressed nor immunotolerized. Animals were sacrificed at 11 weeks after vector injection, and then their brains were assayed for iduronidase activity. Enzyme levels were restored to wild type levels in all parts of the brain, but were significantly lower than in animals that were either immunosuppressed or immunotolerized.
  • FIG. 16 Normalization of glycosaminoglycan (GAG) levels following intracranial or intrathecal AAV9 infusion.
  • AAV9IDUA was injected intracranially or intrathecally into immunodeficient, immunosuppressed or immunotolerized MPS I mice as indicated. Animals were sacrificed 8-11 weeks after injection, then the brains were microdissected and analyzed for GAG levels. GAG storage was restored to wild type levels or close to wild type in all groups analyzed.
  • H hippocampus
  • Cb cerebellum
  • C cortex
  • S striatum
  • ThB thalamus and brain stem.
  • FIGS. 17A-B IDUA vector copies in brain. Microdissected brains were analyzed for IDUA vector sequences by QPCR. The copy numbers in intracranially (A) and intrathecally (B) injected mice correlate to the levels of enzyme activity depicted in FIGS. 11 and 13 .
  • FIG. 18 ICV infusion of AAV8-MCI into adult animals.
  • FIGS. 19A-B Intranasal administration of AAV9/IDUA in immunocompetent, IDUA deficient animals.
  • Adult MPS I mice were infused with AAV9/IDUA intranasally, followed by a weekly immunosuppressive regimen of cyclophosphamide. Animals were sacrificed at 12 weeks post-injection and brains were analyzed for IDUA enzyme activity.
  • FIG. 20 IDUA vector copies in brain. Microdissected brains were analyzed for IDUA vector sequences by QPCR. The copy numbers in intranasally injected mice correlate to the levels of enzyme in FIG. 19 .
  • mammals include, for example, humans; non-human primates, e.g., apes and monkeys; and non-primates, e.g., dogs, cats, rats, mice, cattle, horses, sheep, and goats.
  • Non-mammals include, for example, fish and birds.
  • disease or “disorder” are used interchangeably, and are used to refer to diseases or conditions wherein lack of or reduced amounts of a specific gene product, e.g., a lysosomal storage enzyme, plays a role in the disease such that a therapeutically beneficial effect can be achieved by supplementing, e.g., to at least 1% of normal levels.
  • a specific gene product e.g., a lysosomal storage enzyme
  • “Substantially” as the term is used herein means completely or almost completely; for example, a composition that is “substantially free” of a component either has none of the component or contains such a trace amount that any relevant functional property of the composition is unaffected by the presence of the trace amount, or a compound is “substantially pure” is there are only negligible traces of impurities present.
  • Treating” or “treatment” within the meaning herein refers to an alleviation of symptoms associated with a disorder or disease
  • “inhibiting” means inhibition of further progression or worsening of the symptoms associated with the disorder or disease
  • “preventing” refers to prevention of the symptoms associated with the disorder or disease.
  • an “effective amount” or a “therapeutically effective amount” of an agent of the invention refers to an amount of the agent that alleviates, in whole or in part, symptoms associated with the disorder or condition, or halts or slows further progression or worsening of those symptoms, or prevents or provides prophylaxis for the disorder or condition, e.g., an amount that is effective to prevent, inhibit or treat in the individual one or more neurological symptoms.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of compounds of the invention are outweighed by the therapeutically beneficial effects.
  • a “vector” as used herein refers to a macromolecule or association of macromolecules that comprises or associates with a polynucleotide and which can be used to mediate delivery of the polynucleotide to a cell, either in vitro or in vivo.
  • Illustrative vectors include, for example, plasmids, viral vectors, liposomes and other gene delivery vehicles.
  • the polynucleotide to be delivered sometimes referred to as a “target polynucleotide” or “transgene,” may comprise a coding sequence of interest in gene therapy (such as a gene encoding a protein of therapeutic interest) and/or a selectable or detectable marker.
  • AAV is adeno-associated virus, and may be used to refer to the virus itself or derivatives thereof. The term covers all subtypes, serotypes and pseudotypes, and both naturally occurring and recombinant forms, except where required otherwise.
  • serotype refers to an AAV which is identified by and distinguished from other AAVs based on its binding properties, e.g., there are eleven serotypes of AAVs, AAV-1-AAV-11, including AAV-2, AAV-5, AAV-8, AAV-9 and AAV rh10, and the term encompasses pseudotypes with the same binding properties.
  • AAV-5 serotypes include AAV with the binding properties of AAV-5, e.g., a pseudotyped AAV comprising AAV-5 capsid and a rAAV genome which is not derived or obtained from AAV-5 or which genome is chimeric.
  • rAAV refers to recombinant adeno-associated virus, also referred to as a recombinant AAV vector (or “rAAV vector”).
  • An “AAV virus” refers to a viral particle composed of at least one AAV capsid protein and an encapsidated polynucleotide. If the particle comprises a heterologous polynucleotide (i.e., a polynucleotide other than a wild-type AAV genome such as a transgene to be delivered to a mammalian cell), it is typically referred to as “rAAV”.
  • rAAV heterologous polynucleotide
  • An MV “capsid protein” includes a capsid protein of a wild-type AAV, as well as modified forms of an AAV capsid protein which are structurally and or functionally capable of packaging a rAAV genome and bind to at least one specific cellular receptor which may be different than a receptor employed by wild type AAV.
  • a modified AAV capsid protein includes a chimeric AAV capsid protein such as one having amino acid sequences from two or more serotypes of AAV, e.g., a capsid protein formed from a portion of the capsid protein from AAV-5 fused or linked to a portion of the capsid protein from MV-2, and a AAV capsid protein having a tag or other detectable non-AAV capsid peptide or protein fused or linked to the AAV capsid protein, e.g., a portion of an antibody molecule which binds the transferrin receptor may be recombinantly fused to the AAV-2 capsid protein.
  • a chimeric AAV capsid protein such as one having amino acid sequences from two or more serotypes of AAV, e.g., a capsid protein formed from a portion of the capsid protein from AAV-5 fused or linked to a portion of the capsid protein from MV-2
  • a “pseudotyped” rAAV is an infectious virus having any combination of an AAV capsid protein and an AAV genome.
  • Capsid proteins from any AAV serotype may be employed with a rAAV genome which is derived or obtainable from a wild-type AAV genome of a different serotype or which is a chimeric genome, i.e., formed from AAV DNA from two or more different serotypes, e.g., a chimeric genome having 2 inverted terminal repeats (ITRs), each ITR from a different serotype or chimeric ITRs.
  • ITRs inverted terminal repeats
  • chimeric genomes such as those comprising ITRs from two AAV serotypes or chimeric ITRs can result in directional recombination which may further enhance the production of transcriptionally active intermolecular concatamers.
  • the 5′ and 3′ ITRs within a rAAV vector of the invention may be homologous, i.e., from the same serotype, heterologous, i.e., from different serotypes, or chimeric, i.e., an ITR which has ITR sequences from more than one AAV serotype.
  • Adeno-associated viruses of any serotype are suitable to prepare rAAV, since the various serotypes are functionally and structurally related, even at the genetic level. All AAV serotypes apparently exhibit similar replication properties mediated by homologous rep genes; and all generally bear three related capsid proteins such as those expressed in AAV2. The degree of relatedness is further suggested by heteroduplex analysis which reveals extensive cross-hybridization between serotypes along the length of the genome; and the presence of analogous self-annealing segments at the termini that correspond to ITRs. The similar infectivity patterns also suggest that the replication functions in each serotype are under similar regulatory control. Among the various AAV serotypes, AAV2 is most commonly employed.
  • An AAV vector of the invention typically comprises a polynucleotide that is heterologous to AAV.
  • the polynucleotide is typically of interest because of a capacity to provide a function to a target cell in the context of gene therapy, such as up- or down-regulation of the expression of a certain phenotype.
  • Such a heterologous polynucleotide or “transgene,” generally is of sufficient length to provide the desired function or encoding sequence.
  • heterologous polynucleotide When transcription of the heterologous polynucleotide is desired in the intended target cell, it can be operably linked to its own or to a heterologous promoter, depending for example on the desired level and/or specificity of transcription within the target cell, as is known in the art.
  • a heterologous promoter Various types of promoters and enhancers are suitable for use in this context.
  • Constitutive promoters provide an ongoing level of gene transcription, and may be preferred when it is desired that the therapeutic or prophylactic polynucleotide be expressed on an ongoing basis.
  • Inducible promoters generally exhibit low activity in the absence of the inducer, and are up-regulated in the presence of the inducer.
  • Promoters and enhancers may also be tissue-specific: that is, they exhibit their activity only in certain cell types, presumably due to gene regulatory elements found uniquely in those cells.
  • promoters are the SV40 late promoter from simian virus 40, the Baculovirus polyhedron enhancer/promoter element, Herpes Simplex Virus thymidine kinase (HSV tk), the immediate early promoter from cytomegalovirus (CMV) and various retroviral promoters including LTR elements.
  • Inducible promoters include heavy metal ion inducible promoters (such as the mouse mammary tumor virus (mMTV) promoter or various growth hormone promoters), and the promoters from T7 phage which are active in the presence of T7 RNA polymerase.
  • tissue-specific promoters include various surfactin promoters (for expression in the lung), myosin promoters (for expression in muscle), and albumin promoters (for expression in the liver).
  • surfactin promoters for expression in the lung
  • myosin promoters for expression in muscle
  • albumin promoters for expression in the liver.
  • sequences of many such promoters are available in sequence databases such as the GenBank database.
  • the heterologous polynucleotide will preferably also comprise control elements that facilitate translation (such as a ribosome binding site or “RBS” and a polyadenylation signal).
  • the heterologous polynucleotide generally comprises at least one coding region operatively linked to a suitable promoter, and may also comprise, for example, an operatively linked enhancer, ribosome binding site and poly-A signal.
  • the heterologous polynucleotide may comprise one encoding region, or more than one encoding regions under the control of the same or different promoters. The entire unit, containing a combination of control elements and encoding region, is often referred to as an expression cassette.
  • the heterologous polynucleotide is integrated by recombinant techniques into or in place of the AAV genomic coding region (i.e., in place of the AAV rep and cap genes), but is generally flanked on either side by AAV inverted terminal repeat (ITR) regions.
  • ITR inverted terminal repeat
  • a single ITR may be sufficient to carry out the functions normally associated with configurations comprising two ITRs (see, for example, WO 94/13788), and vector constructs with only one ITR can thus be employed in conjunction with the packaging and production methods of the present invention.
  • the native promoters for rep are self-regulating, and can limit the amount of AAV particles produced.
  • the rep gene can also be operably linked to a heterologous promoter, whether rep is provided as part of the vector construct, or separately. Any heterologous promoter that is not strongly down-regulated by rep gene expression is suitable; but inducible promoters may be preferred because constitutive expression of the rep gene can have a negative impact on the host cell.
  • inducible promoters are known in the art; including, by way of illustration, heavy metal ion inducible promoters (such as metallothionein promoters); steroid hormone inducible promoters (such as the MMTV promoter or growth hormone promoters); and promoters such as those from T7 phage which are active in the presence of T7 RNA polymerase.
  • heavy metal ion inducible promoters such as metallothionein promoters
  • steroid hormone inducible promoters such as the MMTV promoter or growth hormone promoters
  • promoters such as those from T7 phage which are active in the presence of T7 RNA polymerase.
  • T7 RNA polymerase promoters
  • One sub-class of inducible promoters are those that are induced by the helper virus that is used to complement the replication and packaging of the rAAV vector.
  • helper-virus-inducible promoters include the adenovirus early gene promoter which is inducible by adenovirus E1A protein; the adenovirus major late promoter; the herpesvirus promoter which is inducible by herpesvirus proteins such as VP16 or 1CP4; as well as vaccinia or poxvirus inducible promoters.
  • helper-virus-inducible promoters have been described (see, e.g., WO 96/17947). Thus, methods are known in the art to determine whether or not candidate promoters are helper-virus-inducible, and whether or not they will be useful in the generation of high efficiency packaging cells. Briefly, one such method involves replacing the p5 promoter of the MV rep gene with the putative helper-virus-inducible promoter (either known in the art or identified using well-known techniques such as linkage to promoter-less “reporter” genes).
  • the MV rep-cap genes (with p5 replaced), e.g., linked to a positive selectable marker such as an antibiotic resistance gene, are then stably integrated into a suitable host cell (such as the HeLa or A549 cells exemplified below). Cells that are able to grow relatively well under selection conditions (e.g., in the presence of the antibiotic) are then tested for their ability to express the rep and cap genes upon addition of a helper virus. As an initial test for rep and/or cap expression, cells can be readily screened using immunofluorescence to detect Rep and/or Cap proteins. Confirmation of packaging capabilities and efficiencies can then be determined by functional tests for replication and packaging of incoming rAAV vectors.
  • a suitable host cell such as the HeLa or A549 cells exemplified below.
  • helper-virus-inducible promoter derived from the mouse metallothionein gene has been identified as a suitable replacement for the p5 promoter, and used for producing high titers of rAAV particles (as described in WO 96/17947).
  • Removal of one or more AAV genes is in any case desirable, to reduce the likelihood of generating replication-competent AAV (“RCA”). Accordingly, encoding or promoter sequences for rep, cap, or both, may be removed, since the functions provided by these genes can be provided in trans.
  • the resultant vector is referred to as being “defective” in these functions.
  • the missing functions are complemented with a packaging gene, or a plurality thereof, which together encode the necessary functions for the various missing rep and/or cap gene products.
  • the packaging genes or gene cassettes are in one embodiment not flanked by AAV ITRs and in one embodiment do not share any substantial homology with the rAAV genome.
  • the level of homology and corresponding frequency of recombination increase with increasing length of homologous sequences and with their level of shared identity.
  • the level of homology that will pose a concern in a given system can be determined theoretically and confirmed experimentally, as is known in the art. Typically, however, recombination can be substantially reduced or eliminated if the overlapping sequence is less than about a 25 nucleotide sequence if it is at least 80% identical over its entire length, or less than about a 50 nucleotide sequence if it is at least 70% identical over its entire length. Of course, even lower levels of homology are preferable since they will further reduce the likelihood of recombination. It appears that, even without any overlapping homology, there is some residual frequency of generating RCA.
  • the rAAV vector construct, and the complementary packaging gene constructs can be implemented in this invention in a number of different forms.
  • Viral particles, plasmids, and stably transformed host cells can all be used to introduce such constructs into the packaging cell, either transiently or stably.
  • the AAV vector and complementary packaging gene(s), if any, are provided in the form of bacterial plasmids, AAV particles, or any combination thereof.
  • either the AAV vector sequence, the packaging gene(s), or both are provided in the form of genetically altered (preferably inheritably altered) eukaryotic cells. The development of host cells inheritably altered to express the AAV vector sequence, AAV packaging genes, or both, provides an established source of the material that is expressed at a reliable level.
  • a mammalian host cell may be used with at least one intact copy of a stably integrated rAAV vector.
  • An AAV packaging plasmid comprising at least an AAV rep gene operably linked to a promoter can be used to supply replication functions (as described in U.S. Pat. No. 5,658,776).
  • a stable mammalian cell line with an AAV rep gene operably linked to a promoter can be used to supply replication functions (see, e.g., Trempe et al., WO 95/13392); Burstein et al. (WO 98/23018); and Johnson et al. (U.S. Pat.
  • the AAV cap gene providing the encapsidation proteins as described above, can be provided together with an AAV rep gene or separately (see, e.g., the above-referenced applications and patents as well as Allen et al. (WO 98/27204). Other combinations are possible and included within the scope of this invention.
  • Intranasal delivery bypasses the BBB and targets therapeutics directly to the CNS utilizing pathways along olfactory and trigeminal nerves innervating the nasal passages (Frey II, 2002; Thorne et al., 2004; Dhanda et al., 2005).
  • routes of administration to the CNS include intrathecal and intracranial.
  • Intracranial administration may be to the cisterna magna or ventricle.
  • cisterna magna is intended to include access to the space around and below the cerebellum via the opening between the skull and the top of the spine.
  • Cerebral ventricle is intended to include the cavities in the brain that are continuous with the central canal of the spinal cord.
  • Intracranial administration is via injection or infusion and suitable dose ranges for intracranial administration are generally about 10 3 to 10 15 infectious units of viral vector per microliter delivered in 1 to 3000 microliters of single injection volume.
  • viral genomes or infectious units of vector per micro liter would generally contain about 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 , or 10 14 viral genomes or infectious units of viral vector delivered in about 10, 50, 100, 200, 500, 1000, or 2000 microliters.
  • the aforementioned dosage is merely an exemplary dosage and those of skill in the art will understand that this dosage may be varied. Effective doses may be extrapolated from dose-responsive curves derived from in vitro or in vivo test systems.
  • the AAV delivered in the intrathecal methods of treatment of the present invention may be administered through any convenient route commonly used for intrathecal administration.
  • the intrathecal administration may be via a slow infusion of the formulation for about an hour.
  • Intrathecal administration is via injection or infusion and suitable dose ranges for intrathecal administration are generally about 10 3 to 10 15 infectious units of viral vector per microliter delivered in, for example, 1 to 3000 microliters or 0.5 to 15 milliliters of single injection volume.
  • viral genomes or infectious units of vector per microliter would generally contain about 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 , or 10 14 viral genomes or infectious units of viral vector.
  • the therapy results in the normalization of lysosomal storage granules in the neuronal and/or meningeal tissue of the subjects as discussed above. It is contemplated that the deposition of storage granules is ameliorated from neuronal and glial tissue, thereby alleviating the developmental delay and regression seen in individuals suffering with lysosomal storage disease.
  • Other effects of the therapy may include the normalization of lysosomal storage granules in the cerebral meninges near the arachnoid granulation, the presence of which in lysosomal storage disease result in high pressure hydrocephalus.
  • the methods of the invention also may be used in treating spinal cord compression that results from the presence of lysosomal storage granules in the cervical meninges near the cord at C1-05 or elsewhere in the spinal cord.
  • the methods of the invention also are directed to the treatment of cysts that are caused by the perivascular storage of lysosomal storage granules around the vessels of the brain.
  • the therapy also may advantageously result in normalization of liver volume and urinary glycosaminoglycan excretion, reduction in spleen size and apnea/hypopnea events, increase in height and growth velocity in prepubertal subjects, increase in shoulder flexion and elbow and knee extension, and reduction in tricuspid regurgitation or pulmonic regurgitation.
  • the intrathecal administration of the present invention may comprise introducing the composition into the lumbar area. Any such administration may be via a bolus injection. Depending on the severity of the symptoms and the responsiveness of the subject to the therapy, the bolus injection may be administered once per week, once per month, once every 6 months or annually. In other embodiments, the intrathecal administration is achieved by use of an infusion pump. Those of skill in the art are aware of devices that may be used to effect intrathecal administration of a composition.
  • the composition may be intrathecally given, for example, by a single injection, or continuous infusion. It should be understood that the dosage treatment may be in the form of a single dose administration or multiple doses.
  • the term “intrathecal administration” is intended to include delivering a pharmaceutical composition directly into the cerebrospinal fluid of a subject, by techniques including lateral cerebroventricular injection through a burrhole or cistemal or lumbar puncture or the like.
  • the term “lumbar region” is intended to include the area between the third and fourth lumbar (lower back) vertebrae and, more inclusively, the L2-S1 region of the spine.
  • compositions in accordance with the present invention can be achieved by direct injection of the composition or by the use of infusion pumps.
  • the composition can be formulated in liquid solutions, e.g., y in physiologically compatible buffers such as Hank's solution, Ringer's solution or phosphate buffer.
  • the enzyme may be formulated in solid form and re-dissolved or suspended immediately prior to use. Lyophilized forms are also included.
  • the injection can be, for example, in the form of a bolus injection or continuous infusion (e.g., using infusion pumps) of the enzyme.
  • the rAAV is administered by lateral cerebro-ventricular injection into the brain of a subject.
  • the injection can be made, for example, through a burr hole made in the subject's skull.
  • the enzyme and/or other pharmaceutical formulation is administered through a surgically inserted shunt into the cerebral ventricle of a subject.
  • the injection can be made into the lateral ventricles, which are larger, even though injection into the third and fourth smaller ventricles can also be made.
  • the compositions used in the present invention are administered by injection into the cistema magna or lumbar area of a subject.
  • Therapeutics can rapidly gain access to the CNS following intranasal administration along olfactory nerve pathways leading from the nasal cavity directly to the CNS.
  • Olfactory nerve pathways are a major component of intranasal delivery, evidenced by the fact that fluorescent tracers are associated with olfactory nerves as they traverse the cribriform plate (Jansson et al., 2002), drug concentrations in the olfactory bulbs are generally among the highest CNS concentrations observed (Thorne et al., 2004; Banks et al., 2004; Graff et al., 2005a); Nonaka et al., 2008; Ross et al., 2004; Ross et al., 2008; Thorne et al., 2008), and a strong, positive correlation exists between concentrations in the olfactory epithelium and olfactory bulbs (Dhuria et al., 2009a).
  • Olfactory pathways arise in the upper portion of the nasal passages, in the olfactory region, where olfactory receptor neurons (ORNs) are interspersed among supporting cells (sustentacular cells), microvillar cells, and basal cells. ORNs mediate the sense of smell by conveying sensory information from the peripheral environment to the CNS (Clerico et al., 2003). Beneath the epithelium, the lamina basement contains mucus secreting Bowman's glands, axons, blood vessels, lymphatic vessels, and connective tissue.
  • ORNs olfactory receptor neurons
  • the dendrites of ORNs extend into the mucous layer of the olfactory epithelium, while axons of these bipolar neurons extend centrally through the lamina intestinal and through perforations in the cribriform plate of the ethmoid bone, which separates the nasal and cranial cavities.
  • the axons of ORNs pass through the subarachnoid space containing CSF and terminate on mitral cells in the olfactory bulbs. From there, neural projections extend to multiple brain regions including the olfactory tract, anterior olfactory nucleus, piriform cortex, amygdala, and hypothalamus (Buck, 2000).
  • chemosensory neurons located at the anterior tip of the nasal cavity in the Grueneberg ganglion lead into the olfactory bulbs (Fuss et al., 2005; Koos et al., 2005).
  • ORNs The unique characteristics of the ORNs contribute to a dynamic cellular environment critical for intranasal delivery to the CNS. Due to the direct contact with toxins in the external environment, ORNs regenerate every 3-4 weeks from basal cells residing in the olfactory epithelium (Mackay-Sim, 2003). Special Schwann cell-like cells called olfactory ensheathing cells (OECs) envelope the axons of ORNs and have an important role in axonal regeneration, regrowth, and remyelination (Field et al., 2003; Li et al., 2005a; Li et al., 2005b). The OECs create continuous, fluid-filled perineurial channels that, interestingly, remain open, despite the degeneration and regeneration of ORNs (Williams et al., 2004).
  • OECs olfactory ensheathing cells
  • Extracellular transport mechanisms involve the rapid movement of molecules between cells in the nasal epithelium, requiring only several minutes to 30 minutes for a drug to reach the olfactory bulbs and other areas of the CNS after intranasal administration (Frey II, 2002; Balin et al., 1986). Transport likely involves bulk flow mechanisms (Thorne et al., 2004; Thorne et al., 2001) within the channels created by the OECs.
  • Drugs may also be propelled within these channels by the structural changes that occur during depolarization and axonal propagation of the action potential in adjacent axons (Luzzati et al., 2004).
  • Intracellular transport mechanisms involve the uptake of molecules into ORNs by passive diffusion, receptor-mediated endocytosis or adsorptive endocytosis, followed by slower axonal transport, taking several hours to days for a drug to appear in the olfactory bulbs and other brain areas (Baker et al., 1986; Broadwell et al., 1985; Kristensson et al., 1971).
  • Intracellular transport in ORNs has been demonstrated for small, lipophilic molecules such as gold particles (de Lorenzo, 1970; Gopinath et al., 1978), aluminum salts (Perl et al., 1987), and for substances with receptors on ORNs such as WGA-HRP (Thorne et al., 1995; Baker et al., 1986; Itaya et al., 1986; Shipley, 1985). Intracellular mechanisms, while important for certain therapeutics, are not likely to be the predominant mode of transport into the CNS.
  • GLP galanin-like peptide
  • IGF-I insulin-like growth factor-I
  • the trigeminal nerve conveys sensory information from the nasal cavity, the oral cavity, the eyelids, and the cornea, to the CNS via the ophthalmic division (V1), the maxillary division (V2), or the mandibular division (V3) of the trigeminal nerve (Clerico et al., 2003; Gray, 1978).
  • V1 the ophthalmic division
  • V2 the maxillary division
  • V3 the mandibular division
  • Branches from the ophthalmic division of the trigeminal nerve provide innervation to the dorsal nasal mucosa and the anterior portion of the nose, while branches of the maxillary division provide innervation to the lateral walls of the nasal mucosa.
  • the mandibular division of the trigeminal nerve extends to the lower jaw and teeth, with no direct neural inputs to the nasal cavity.
  • the three branches of the trigeminal nerve come together at the trigeminal ganglion and extend centrally to enter the brain at the level of the pons, terminating in the spinal trigeminal nuclei in the brainstem.
  • a unique feature of the trigeminal nerve is that it enters the brain from the respiratory epithelium of the nasal passages at two sites: (1) through the anterior lacerated foramen near the pons and (2) through the cribriform plate near the olfactory bulbs, creating entry points into both caudal and rostral brain areas following intranasal administration. It is also likely that other nerves that innervate the face and head, such as the facial nerve, or other sensory structures in the nasal cavity, such as the Grueneberg ganglion, may provide entry points for intranasally applied therapeutics into the CNS.
  • the intranasal route of administration has been utilized to deliver drugs to the systemic circulation via absorption into the capillary blood vessels underlying the nasal mucosa.
  • the nasal mucosa is highly vascular, receiving its blood supply from branches of the maxillary, ophthalmic and facial arteries, which arise from the carotid artery (Clerico et al., 2003; Cauna, 1982).
  • the olfactory mucosa receives blood from small branches of the ophthalmic artery, whereas the respiratory mucosa receives blood from a large caliber arterial branch of the maxillary artery (DeSesso, 1993).
  • the relative density of blood vessels is greater in the respiratory mucosa compared to the olfactory mucosa, making the former region an ideal site for absorption into the blood (DeSesso, 1993).
  • the vasculature in the respiratory region contains a mix of continuous and fenestrated endothelia (Grevers et al., 1987; Van Diest et al., 1979), allowing both small and large molecules to enter the systemic circulation following nasal administration.
  • Perivascular spaces are bound by the outermost layer of blood vessels and the basement membrane of the surrounding tissue (Pollock et al., 1997). These perivascular spaces act as a lymphatic system for the brain, where neuron-derived substances are cleared from brain interstitial fluid by entering perivascular channels associated with cerebral blood vessels.
  • Perivascular transport is due to bulk flow mechanisms, as opposed to diffusion alone (Cserr et al., 1981; Groothuis et al., 2007), and arterial pulsations are also a driving force for perivascular transport (Rennels et al., 1985; Rennels et al., 1985). Intranasally applied drugs can move into perivascular spaces in the nasal passages or after reaching the brain and the widespread distribution observed within the CNS could be due to perivascular transport mechanisms (Thorne et al., 2004).
  • Pathways connecting the subarachnoid space containing CSF, perineurial spaces encompassing olfactory nerves, and the nasal lymphatics are important for CSF drainage and these same pathways provide access for intranasally applied therapeutics to the CSF and other areas of the CNS.
  • Optimal delivery to the CNS along neural pathways is associated with delivery of the agent to the upper third of the nasal cavity (Hanson et al., 2008).
  • a supine position may be employed another position for targeting the olfactory region is with the “praying to Mecca” position, with the head down-and-forward.
  • a supine position with the head angle at 70° or 90° may be suitable for efficient delivery to the CSF using a tube inserted into the nostrils to deliver the drug via intranasal administration (van den Berg et al., (2002)).
  • nose drops may be administered over a period of 10-20 minutes to alternating nostrils every 1-2 minutes to allow the solution to be absorbed into the nasal epithelium (Thorne et al., 2004; Capsoni et al., 2002; Ross et al., 2004; Ross et al., 2008; Dhuria et al., 2009a; Dhuria et al., 2009b; Francis et al., 2008; Martinez et al., 2008).
  • This noninvasive method does not involve inserting the device into the nostril. Instead, drops are placed at the opening of the nostril, allowing the individual to sniff the drop into the nasal cavity.
  • Flexible tubing can be inserted into the nostrils for localized delivery of a small volume of the drug solution to the respiratory or olfactory epithelia, depending on the length of the tubing (Chow et al., 1999; Van den Berg et al., 2003; van den Berg et al., 2004a; Banks et al., 2004; van den Berg et al., 2002; Vyas et al., 2006a; Charlton et al., 2007a; Gao et al., 2007a).
  • Nasal delivery devices such as sprays, nose droppers or needle-less syringes, may be employed to target the agent to different regions of the nasal cavity.
  • OptiMistTM is a breath actuated device that targets liquid or powder nasal formulations to the nasal cavity, including the olfactory region, without deposition in the lungs or esophagus (Djupesland et al., 2006).
  • the ViaNaseTM device can also be used to target a nasal spray to the olfactory and respiratory epithelia of the nasal cavity.
  • Nasal drops tend to deposit on the nasal floor and are subjected to rapid mucociliary clearance, while nasal sprays are distributed to the middle meatus of the nasal mucosa (Scheibe et al., 2008).
  • the immune suppressant or immunotolerizing agent may be administered by any route including parenterally.
  • the immune suppressant or immunotolerizing agent may be administered by subcutaneous, intramuscular, or intravenous injection, orally, intrathecally, intracranially, or intranasally, or by sustained release, e.g., using a subcutaneous implant.
  • the immune suppressant or immunotolerizing agent may be dissolved or dispersed in a liquid carrier vehicle.
  • the active material may be suitably admixed with an acceptable vehicle, e.g., of the vegetable oil variety such as peanut oil, cottonseed oil and the like.
  • compositions may comprise an aqueous solution of a water soluble pharmaceutically acceptable salt of the active acids according to the invention, desirably in a concentration of 0.01-10%, and optionally also a stabilizing agent and/or buffer substances in aqueous solution. Dosage units of the solution may advantageously be enclosed in ampules.
  • the composition e.g., rAAV containing composition, immune suppressant containing composition or immunotolerizing composition
  • carriers or diluents usable for preparing such injectable doses include diluents such as water, ethyl alcohol, macrogol, propylene glycol, ethoxylated isostearyl alcohol, polyoxyisostearyl alcohol and polyoxyethylene sorbitan fatty acid esters, pH adjusting agents or buffers such as sodium citrate, sodium acetate and sodium phosphate, stabilizers such as sodium pyrosulfite, EDTA, thioglycolic acid and thiolactic acid, isotonic agents such as sodium chloride and glucose, local anesthetics such as procaine hydrochloride and lidocaine hydrochloride.
  • injections can be prepared by adding such carriers to the enzyme or other active, following procedures well known to those of skill in the art.
  • a thorough discussion of pharmaceutically acceptable excipients is available in REMINGTON'S PHARMACEUTICAL SCIENCES (Mack Pub. Co., N.J. 1991).
  • the pharmaceutically acceptable formulations can easily be suspended in aqueous vehicles and introduced through conventional hypodermic needles or using infusion pumps. Prior to introduction, the formulations can be sterilized with, preferably, gamma radiation or electron beam sterilization.
  • the immune suppressant or immunotolerizing agent When the immune suppressant or immunotolerizing agent is administered in the form of a subcutaneous implant, the compound is suspended or dissolved in a slowly dispersed material known to those skilled in the art, or administered in a device which slowly releases the active material through the use of a constant driving force such as an osmotic pump. In such cases, administration over an extended period of time is possible.
  • the dosage at which the immune suppressant or immunotolerizing agent containing composition is administered may vary within a wide range and will depend on various factors such as the severity of the disease, the age of the patient, etc., and may have to be individually adjusted.
  • a possible range for the amount which may be administered per day is about 0.1 mg to about 2000 mg or about 1 mg to about 2000 mg.
  • the compositions containing the immune suppressant or immunotolerizing agent may suitably be formulated so that they provide doses within these ranges, either as single dosage units or as multiple dosage units.
  • the subject formulations may contain one or more rAAV encoding a therapeutic gene product.
  • compositions described herein may be employed in combination with another medicament.
  • the compositions can appear in conventional forms, for example, aerosols, solutions, suspensions, or topical applications, or in lyophilized form.
  • compositions include a rAAV, an immune suppressant, a permeation enhancer, or a combination thereof, and a pharmaceutically acceptable excipient which can be a carrier or a diluent.
  • the active agent(s) may be mixed with a carrier, or diluted by a carrier, or enclosed within a carrier.
  • the active agent when the active agent is mixed with a carrier, or when the carrier serves as a diluent, it can be solid, semi-solid, or liquid material that acts as a vehicle, excipient, or medium for the active agent.
  • suitable carriers are water, salt solutions, alcohols, polyethylene glycols, polyhydroxyethoxylated castor oil, peanut oil, olive oil, gelatin, lactose, terra alba, sucrose, dextrin, magnesium carbonate, sugar, cyclodextrin, amylose, magnesium stearate, talc, gelatin, agar, pectin, acacia, stearic acid or lower alkyl ethers of cellulose, silicic acid, fatty acids, fatty acid amines, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, polyoxyethylene, hydroxymethylcellulose and polyvinylpyrrolidone.
  • the carrier or diluent can include any sustained release material known in the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax.
  • the formulations can be mixed with auxiliary agents which do not deleteriously react with the active agent(s).
  • auxiliary agents which do not deleteriously react with the active agent(s).
  • Such additives can include wetting agents, emulsifying and suspending agents, salt for influencing osmotic pressure, buffers and/or coloring substances preserving agents, sweetening agents or flavoring agents.
  • the compositions can also be sterilized if desired.
  • the preparation can be in the form of a liquid such as an aqueous liquid suspension or solution.
  • Acceptable solvents or vehicles include sterilized water, Ringer's solution, or an isotonic aqueous saline solution.
  • the agent(s) may be provided as a powder suitable for reconstitution with an appropriate solution as described above. Examples of these include, but are not limited to, freeze dried, rotary dried or spray dried powders, amorphous powders, granules, precipitates, or particulates.
  • the composition can optionally contain stabilizers, pH modifiers, surfactants, bioavailability modifiers and combinations of these.
  • a unit dosage form can be in individual containers or in multi-dose containers.
  • compositions contemplated by the present invention may include, for example, micelles or liposomes, or some other encapsulated form, or can be administered in an extended release form to provide a prolonged storage and/or delivery effect, e.g., using biodegradable polymers, e.g., polylactide-polyglycolide.
  • biodegradable polymers e.g., polylactide-polyglycolide.
  • examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Polymeric nanoparticles e.g., comprised of a hydrophobic core of polylactic acid (PLA) and a hydrophilic shell of methoxy-poly(ethylene glycol) (MPEG), may have improved solubility and targeting to the CNS. Regional differences in targeting between the microemulsion and nanoparticle formulations may be due to differences in particle size.
  • PLA polylactic acid
  • MPEG methoxy-poly(ethylene glycol)
  • Liposomes are very simple structures consisting of one or more lipid bilayers of amphiphilic lipids, i.e., phospholipids or cholesterol. The lipophilic moiety of the bilayers is turned towards each other and creates an inner hydrophobic environment in the membrane. Liposomes are suitable drug carriers for some lipophilic drugs which can be associated with the non-polar parts of lipid bilayers if they fit in size and geometry. The size of liposomes varies from 20 nm to few ⁇ m.
  • Mixed micelles are efficient detergent structures which are composed of bile salts, phospholipids, tri, di- and monoglycerides, fatty acids, free cholesterol and fat soluble micronutrients.
  • long-chain phospholipids are known to form bilayers when dispersed in water
  • the preferred phase of short chain analogues is the spherical micellar phase.
  • a micellar solution is a thermodynamically stable system formed spontaneously in water and organic solvents.
  • the interaction between micelles and hydrophobic/lipophilic drugs leads to the formation of mixed micelles (MM), often called swallen micelles, too.
  • MM mixed micelles
  • Lipid microparticles includes lipid nano- and microspheres.
  • Microspheres are generally defined as small spherical particles made of any material which are sized from about 0.2 to 100 ⁇ m. Smaller spheres below 200 nm are usually called nanospheres.
  • Lipid microspheres are homogeneous oil/water microemulsions similar to commercially available fat emulsions, and are prepared by an intensive sonication procedure or high pressure emulsifying methods (grinding methods). The natural surfactant lecithin lowers the surface tension of the liquid, thus acting as an emulsifier to form a stable emulsion.
  • the structure and composition of lipid nanospheres is similar to those of lipid microspheres, but with a smaller diameter.
  • Polymeric nanoparticles serve as carriers for a broad variety of ingredients.
  • the active components may be either dissolved in the polymetric matrix or entrapped or adsorbed onto the particle surface.
  • Polymers suitable for the preparation of organic nanoparticles include cellulose derivatives and polyesters such as poly(lactic acid), poly(glycolic acid) and their copolymer. Due to their small size, their large surface area/volume ratio and the possibility of functionalization of the interface, polymeric nanoparticles are ideal carrier and release systems. If the particle size is below 50 nm, they are no longer recognized as particles by many biological and also synthetic barrier layers, but act similar to molecularly disperse systems.
  • composition of the invention can be formulated to provide quick, sustained, controlled, or delayed release, or any combination thereof, of the active agent after administration to the individual by employing procedures well known in the art.
  • the enzyme is in an isotonic or hypotonic solution.
  • a lipid based delivery vehicle may be employed, e.g., a microemulsion such as that described in WO 2008/049588, the disclosure of which is incorporated by reference herein, or liposomes.
  • the preparation can contain an agent, dissolved or suspended in a liquid carrier, such as an aqueous carrier, for aerosol application.
  • a liquid carrier such as an aqueous carrier
  • the carrier can contain additives such as solubilizing agents, e.g., propylene glycol, surfactants, absorption enhancers such as lecithin (phosphatidylcholine) or cyclodextrin, or preservatives such as parabens.
  • solubilizing agents e.g., propylene glycol
  • surfactants e.g., surfactants
  • absorption enhancers such as lecithin (phosphatidylcholine) or cyclodextrin
  • preservatives such as parabens.
  • efficient delivery to the CNS following intranasal administration may be dependent on membrane permeability.
  • improving membrane permeability may enhance extracellular mechanisms of transport to the CNS along olfactory and trigeminal nerves.
  • permeation enhancers such as surfactants, e.g., lauroylcarnitine (LC), bile salts, lipids, cyclodextrins, polymers, or tight junction modifiers.
  • the active agents are dispensed in unit dosage form including the active ingredient together with a pharmaceutically acceptable carrier per unit dosage.
  • dosage forms suitable for nasal administration include from about 125 lug to about 125 mg, e.g., from about 250 lug to about 50 mg, or from about 2.5 mg to about 25 mg, of the compounds admixed with a pharmaceutically acceptable carrier or diluent.
  • Dosage forms can be administered daily, or more than once a day, such as twice or thrice daily. Alternatively dosage forms can be administered less frequently than daily, such as every other day, or weekly, if found to be advisable by a prescribing physician.
  • MPS I Mucopolysaccharidosis type I
  • IDUA alpha-L-iduronidase
  • Sic and abnormal accumulation of glycosaminoglycans is associated with growth delay, organomegaly, skeletal dysplasia, and cardiopulmonary disease.
  • Individuals with the most severe form of the disease suffer from neurodegeneration, mental retardation, and early death.
  • the two current treatments for MPS I hematopoietic stem cell transplantation and enzyme replacement therapy
  • CNS central nervous system
  • AAV9-IDUA preparation The AAV-IDUA vector construct (MCI) has been previously described (Wolf et al., 2011) (mCags promoter). AAV-IDUA plasmid DNA was packaged into AAV9 virions at the University of Florida Vector Core, yielding a titer of 3 ⁇ 10 13 vector genomes per milliliter.
  • ICV infusions Adult Idua ⁇ / ⁇ mice were anesthetized using a cocktail of ketamine and xylazine (100 mg ketamine+10 mg xylazine per kg) and placed on a stereotactic frame. Ten microliters of AAV9-IDUA were infused into the right-side lateral ventricle (stereotactic coordinates AP 0.4, ML 0.8, DV 2.4 mm from bregma) using a Hamilton syringe. The animals were returned to their cages on heating pads for recovery.
  • Intrathecal infusions were carried out by injection of 10 ⁇ L AAV vector containing solution between the L5 and L6 vertebrae 20 minutes after intravenous injection of 0.2 mL 25% mannitol.
  • Newborn IDUA deficient mice were injected through the facial temporal vein with 5 ⁇ L containing 5.8 ⁇ g of recombinant iduronidase protein (Aldurazyme), and then the animals were returned to their cage.
  • Aldurazyme recombinant iduronidase protein
  • Cyclophosphamide immunosuppression animals were administered cyclophosphamide once per week at a dose of 120 mg/kg starting one day after infusion with AAV9-IDUA vector.
  • Tissues Tissue homogenates were clarified by centrifugation for 3 minutes at 13,000 rpm using an Eppendorf tabletop centrifuge model 5415D (Eppendorf) and incubated overnight with proteinase K, DNase1, and Rnase. GAG concentration was determined using the Blyscan Sulfated Glycosaminoglycan Assay (Accurate Chemical) according to the manufacturer's instructions.
  • FIG. 1 shows the results for iduronidase-deficient mice that were administered MV either intracerebroventricularly (ICV) or intrathecally (IT).
  • MV intracerebroventricularly
  • IT intrathecally
  • animals were either immunosuppressed with cyclophosphamide (CP), immunotolerized at birth by intravenous administration of human iduonidase protein (aldurazyme), or the injections were carried out in NOD-SCID immunodeficient mice that were also iduronidase deficient. Animals were sacrificed at the indicated time post-treatment, the brains were microdissected and extracts assayed for iduronidase activity.
  • CP cyclophosphamide
  • aldurazyme human iduonidase protein
  • FIG. 2 illustrates data for immunodeficient, IDUA deficient animals injected ICV with AAV-IDUA vector. Those animals exhibited high levels of IDUA expression (10 to 100 times wild type) in all areas of the brain, with the highest level observed in the brain stem and thalamus (“rest”).
  • Immunosuppressed animals administered AAV vector by ICV route had a relatively lower level of enzyme in the brain compared to the immunodeficient animals ( FIG. 3 ). Note that immunosuppression may have been compromised in these animals because CP was withdrawn 2 weeks before sacrifice due to poor health.
  • FIG. 4 shows data for immunosuppressed animals administered AAV vector by the IT route.
  • Immunotolerized animals administered AAV vector ICV exhibited widespread IDUA activity in all parts of the brain ( FIG. 5 ), similar to that observed in the immunodeficient animals, indicating the effectiveness of the immunotolerization procedure.
  • FIG. 6 is a compilation of all mean levels of IDUA activity for side-by-side comparison
  • FIG. 7 is data grouped according the area of the brain.
  • GAG storage material was assayed in the different sections of the brain for all four of the test groups. For each group, the mean of each portion of the brain is shown on the left, the values for each of the individual animals is shown on the right ( FIG. 8 ). IDUA deficient animals (far left) contained high levels of GAG compared to wild type animals (magenta bar). GAG levels were at wild-type or lower than wild type for all portions of the brain in all groups of AAV-treated animals. GAG levels were slightly although not significantly higher than wild-type in cortex and brainstem of animals administered AAV9-IDUA intrathecally.
  • AAV9IDUA Preparation.
  • AAV-IDUA plasmid was packaged into AAV9 virions at either the University of Florida vector core, or the University of Pennsylvania vector core, yielding a titer of 1-3 ⁇ 10 13 vector genomes per millilter.
  • Animals Animals. Animals were anesthetized with ketamine/xylazine (100 mg ketamine+10 mg xylazine per kg) and transcardially perfused with 70 mL PBS prior to sacrifice. Brains were harvested and microdissected on ice into cerebellum, hippocampus, striatum, cortex, and brainstem/thalamus (“rest”). The samples were frozen on dry ice and then stored at ⁇ 80° C.
  • Tissue IDUA activity Tissue samples were thawed and homogenized in saline in a tissue homogenizer. Tissue homogenates were clarified by centrifugation at 15,000 rpm in a benchtop Eppendorf centrifuge at 4° C. for 15 minutes. Tissue lysates (supernatant) were collected and analyzed for IDUA activity and GAG storage levels.
  • Tissue GAG levels Tissue lysates were incubated overnight with Proteinase K, RNase and DNase. GAG levels were analyzed using the Blyscan Sulfated Glycosaminoglycan Assay according to the manufacturer's instructions.
  • Tissue homogenates were used for DNA isolation and subsequent QPCR, as described in Wolf et al. (2011).
  • FIG. 9 illustrates the experimental design and groups.
  • Animals were administered AAV9IDUA vector either by intracerebroventricular (ICV) or intrathecal (IT) infusion.
  • Vector administration was carried out in NOD-SCID immunodeficient (ID) mice that were also IDUA deficient, or in IDUA deficient mice that were either immunosuppressed with cyclophosphamide (CP), or immunotolerized at birth by a single or multiple injections of human iduronidase protein (Aldurazyme). The times of treatment with vector and sacrifice are as indicated in FIG. 9 .
  • All vector administrations were carried out in adult animals ranging in age from 3-4.5 months. Animals were injected with 10 ⁇ L of vector at a dose of 3 ⁇ 10 11 vector genomes per 10 microliters.
  • FIG. 10 shows IDUA enzyme activities in intracranially infused, immunodeficient, IDUA deficient mice. High levels of enzyme activity were seen in all areas of the brain, ranging from 30- to 300-fold higher than wild type levels. Highest enzyme expressions were seen in thalamus and brain stem, and in the hippocampus.
  • CP cyclophosphamide
  • IDUA enzyme levels in animals tolerized at birth with IDUA protein (Aldurazyme) and administered vector intracranially are depicted in FIG. 12 . All animals showed high enzyme levels in all parts of the brain that ranged from 10- to 1000-fold higher than wild type levels, similar to levels achieved in immunodeficient animals, indicating the effectiveness of the immunotolerization procedure.
  • FIG. 13 depicts IDUA enzyme levels in mice that were injected intrathecally and administered CP on a weekly basis. Elevated levels of IDUA were observed in all parts of the brain, especially in the cerebellum and the spinal cord. Levels of enzyme were the lowest in the striatum and hippocampus with activities at wild type levels.
  • IDUA deficient mice were tolerized with Aldurazyme as described, and injected with vector intrathecally ( FIG. 14 ). There was widespread IDUA enzyme activity in all parts of the brain, with highest levels of activity in the brain stem and thalamus, olfactory bulb, spinal cord and the cerebellum. Similar to the data in FIG. 13 , the lowest levels of enzyme activity were seen in the striatum, cortex and hippocampus.
  • AAV9IDUA vector in animals that were immunotolerized and injected with vector either intracranially or intrathecally was evaluated by QPCR, as illustrated in FIG. 16 .
  • IDUA copies per cell were higher in animals infused intracranially in comparison with animals infused intrathecally, which is consistent with the higher level of enzyme activity seen in animals injected intracranially.
  • IDUA High, widespread, and therapeutic levels of IDUA were observed in all areas of the brain after intracerebroventricular and intrathecal routes of AAV9IDUA administration in adult mice Enzyme activities were restored to wild type levels or slightly higher in immunocompetent IDUA deficient animals infused with AAV-IDUA intrathecally. Significantly higher levels of IDUA enzyme were observed for both routes of vector injection in animals immunotolerized starting at birth by administration of IDUA protein.
  • mice (12 weeks old) were anesthetized with ketamine/xylazine, followed by intranasal infusion of AAV9IDUA vector.
  • Vector was administered by applying eight 3 ⁇ L drops with a micropipette to the intranasal cavity, alternating between nostrils, at 2 minute intervals between each application.
  • a total of 2.4-7 ⁇ 10 11 vector genomes was administered to each adult animal, depending on source of vector.
  • Animals were immunosuppressed with 120 mg/kg cyclophosphamide administered weekly, starting the day after vector administration. Mice were sacrificed at 12 weeks post vector infusion, animals were assayed for IDUA enzyme expression and vector copies in the brain.

Abstract

A method to prevent, inhibit or treat one or more symptoms associated with a disease of the central nervous system by intrathecally, intracerebroventricularly or endovascularly administering a rAAV encoding a gene product associated with the disease, e.g., a mammal in which the gene product is absent or present at a reduced level relative to a mammal without the disease.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 15/995,045, filed May 31, 2018, which is a divisional of U.S. patent application Ser. No. 15/717,450, filed Sep. 27, 2017, which is a continuation of U.S. patent application Ser. No. 14/889,750, filed Nov. 6, 2015, now U.S. Pat. No. 9,827,295, which is a U.S. National Stage filing under 35 U.S.C. 371 from International Application No. PCT/US2014/038209, filed on May 15, 2014 and published as WO 2014/186579 A1 on 20 Nov. 2014, which claims the benefit of the filing date of U.S. application Ser. No. 61/823,757, filed on May 15, 2013, the disclosures of which are incorporated by reference herein.
  • STATEMENT OF GOVERNMENT RIGHTS
  • This invention was made with government support under HD032652 and DK094538 awarded by National Institutes of Health. The Government has certain rights in the invention.
  • BACKGROUND
  • The mucopolysaccharidoses (MPSs) are a group of 11 storage diseases caused by disruptions in glycosaminoglycan (GAG) catabolism, leading to their accumulation in lysosomes (Muenzer, 2004; Munoz-Rojas et al., 2008). Manifestations of varying severity include organomegaly, skeletal dysplasias, cardiac and pulmonary obstruction and neurological deterioration. For MPS I, deficiency of iduronidase (IDUA), severity ranges from mild (Scheie syndrome) to moderate (Hurler-Scheie) to severe (Hurler syndrome), with the latter resulting in neurologic deficiency and death by age 15 (Muenzer, 2004; Munoz-Rojas et al., 2008). Therapies for MPSs have been for the most part palliative. However, there are some of the MPS diseases, including Hurler syndrome, for which allogeneic hematopoietic stem cell transplantation (HSCT) has exhibited efficacy (Krivit, 2004; Orchard et al., 2007; Peters et al., 2003). Additionally, for more and more of the MPS diseases, enzyme replacement therapy (ERT) is becoming available (Brady, 2006). In general, HSCT and ERT result in the clearing of storage materials and improved peripheral conditions, although some problems persist after treatment (skeletal, cardiac, corneal clouding). The primary challenge in these cellular and enzyme therapies is effectiveness in addressing neurological manifestations, as peripherally administered enzyme does not penetrate the blood-brain barrier and HSCT has been found to be of benefit for some, but not all, MPS's.
  • MPS I has been one of the most extensively studied of the MPS diseases for development of cellular and molecular therapies. The effectiveness of allogeneic HSCT is most likely the result of metabolic cross-correction, whereby the missing enzyme is released from donor-derived cells and subsequently taken up by host cells and trafficked to lysosomes, where the enzyme contributes to lysosomal metabolism (Fratantoni et al., 1968). Clearing of GAG storage materials is subsequently observed in peripheral organs such as liver and spleen, there is relief from cardiopulmonary obstruction and improvement in corneal clouding (Orchard et al., 2007). Of particular importance is the effect of allogeneic stem cell transplantation on the emergence of neurologic manifestations in the MPS diseases. In this regard, there is evidence for several MPS diseases that individuals engrafted with allogeneic stem cells face an improved outcome in comparison with untransplanted patients (Bjoraker et al., 2006; Krivit, 2004; Orchard et al., 2007; Peters et al., 2003). A central hypothesis explaining the neurologic benefit of allogeneic hematopoietic stem cell transplant is the penetration of donor-derived hematopoietic cells (most likely microglia) (Hess et al., 2004; Unger et al., 1993) into the central nervous system, where the missing enzyme is expressed by engrafted cells from which point the enzyme diffuses into CNS tissues and participates in clearing of storage materials. The level of enzyme provided to CNS tissues is thus limited to that amount expressed and released from donor-derived cells engrafting in the brain. While such engraftment is of great benefit for MPS I, recipients nonetheless continue to exhibit below normal IQ and impaired neurocognitive capability (Ziegler and Shapiro, 2007).
  • The phenomenon of metabolic cross correction also explains the effectiveness of ERT for several lysosomal storage diseases (Brady, 2006), most notably MPS I. However, due to the requirement for penetration of the blood-brain barrier (BBB) by the enzyme missing in the particular lysosomal storage disease (LSD) in order to effectively reach the CNS, effectiveness of enzyme therapy for neurologic manifestations of lysosomal storage disease (LSD) has not been observed (Brady, 2006). Enzymes are almost always too large and generally too charged to effectively cross the BBB. This has prompted investigations into invasive intrathecal enzyme administration (Dickson et al., 2007), for which effectiveness has been demonstrated in a canine model of MPS I (Kakkis et al., 2004) and for which human clinical trials are beginning for MPS I (Pastores, 2008; Munoz-Rojas et al., 2008). Key disadvantages of enzyme therapy include its great expense (>$200,000 per year) and the requirement for repeated infusions of recombinant protein. Current clinical trials of intrathecal IDUA administration are designed to inject the enzyme only once every three months, so the effectiveness of this dosing regimen remains uncertain.
  • SUMMARY OF THE INVENTION
  • Methods of preventing, inhibiting, and/or treating one or more symptoms associated with a disease of the central nervous system (CNS) in a mammal in need thereof are described. The methods involve delivering to the CNS of a mammal in need of treatment a composition comprising an effective amount of a recombinant adeno-associated virus (rAAV) vector comprising an open reading frame encoding a gene product, e.g., a therapeutic gene product. Target gene products that may be encoded by an rAAV vector include, but are not limited to, alpha-L-iduronidase, iduronate-2-sulfatase, heparan sulfate sulfatase, N-acetyl-alpha-D-glucosaminidase, beta-hexosaminidase, alpha-galactosidase, betagalactosidase, beta-glucuronidase or glucocerebrosidase. Diseases that may be prevented, inhibited or treated using the methods disclosed herein include, but are not limited to, mucopolysaccharidosis type I disorder, a mucopolysaccharidosis type II disorder, or a mucopolysaccharidosis type VII disorder. The AAV vector can be administered in a variety of ways to ensure that it is delivered to the CNS/brain, and that the transgene is successfully transduced in the subject's CNS/brain. Routes of delivery to the CNS/brain include, but are not limited to intrathecal administration, intracranial administration, e.g., intracerebroventricular administration, or lateral cerebro ventricular administration), intranasal administration, endovascular administration, and intraparenchymal administration.
  • In one embodiment, the methods involve delivering to the CNS of an adult mammal in need of treatment a composition comprising an effective amount of a rAAV-9 vector comprising an open reading frame encoding a gene. In one embodiment, the methods involve delivering to the CNS of an adult mammal in need of treatment a composition comprising an effective amount of a rAAV-9 vector comprising an open reading frame encoding IDUA. These methods are based, in part, on the discovery that an AAV-9 vector can efficiently transduce the therapeutic transgene in the brain/CNS of adult subjects, restoring enzyme levels to wild type levels. (see FIG. 15, infra). The results achieved using AAV-9 are surprising in view of previous work which demonstrated that intravascular delivery of AAV-9 in adult mice does not achieve widespread direct neuronal targeting (see Foust et al., 2009), as well as additional data showing that direct injection of AAV8-IDUA into the CNS of adult IDUA-deficient mice resulted in poor transgene expression (FIG. 18). As proof of principle, the working examples described herein use a pre-clinical model for the treatment of MPS1, an inherited metabolic disorder caused by deficiency of the lysosomal enzyme alpha-L-iduronidase (IDUA). The working examples surprisingly demonstrate that direct injection of AAV9-IDUA into the CNS of immunocompetent adult IDUA-deficient mice resulted in IDUA enzyme expression and activity that is the same or higher than IDUA enzyme expression and activity in wild-type adult mice (see FIG. 15, infra).
  • In an additional embodiment of the invention, the working examples also demonstrate that co-therapy to induce immunosuppression or immunotolerization, or treatment of immunodeficient animals, can achieve even higher levels of IDUA enzyme expression and activity. In an embodiment, patients with genotypes that promote an immune response that neutralizes enzyme activity (see, e.g., Barbier et al., 2013) are treated with an immunosuppressant in addition to the rAAV vector comprising an open reading frame encoding a gene product, such as IDUA.
  • Neonatal IDUA−/− mice are not immunocompetent. However administration of IDUA expressing AAV-8 to neonatal IDUA−/− mice resulted in IDUA expression (Wolf et al., 2011), thus tolerizing the animals to IDUA. As described herein, the applicability of AAV-mediated gene transfer to adult (immunocompetent) mice by direct infusion of AAV to the central nervous system was shown using different routes of administration. For example, AAV-IDUA serotype 9 was administered by direct injection into the lateral ventricles of adult IDUA-deficient mice that were either immunocompetent, immunodeficient (NODSCID/IDUA−/−), immunosuppressed with cyclophosphamide (CP), or immunotolerized by weekly injection of human iduronidase protein (Aldurazyme) starting at birth. CP immunosuppressed animals were also administered AAV9-IDUA by intranasal infusion, by intrathecal injection, and by endovascular infusion with and without mannitol to disrupt the blood-brain barrier. Animals were sacrificed at 8 weeks after vector administration, and brains were harvested and microdissected for evaluation of IDUA enzymatic activity, tissue glycosaminoglycans, and IDUA vector sequences in comparison with normal and affected control mice. Results from these studies show that numerous routes for AAV vector administration directly to the CNS may be employed, e.g., so as to achieve higher levels of protein delivery and/or enzyme activity in the CNS. In addition, although the brain is an immunopriviledged site, administration of an immunosuppressant or immunotolerization may increase the activity found in the brain after AAV administration. Higher levels of expression per administration and/or less invasive routes of administration are clinically more palatable to patients.
  • Thus, the invention includes the use of recombinant AAV (rAAV) vectors that encode a gene product with therapeutic effects when expressed in the CNS of a mammal. In one embodiment, the mammal is an immunocompetent mammal with a disease or disorder of the CNS (a neurologic disease). An “immunocompetent” mammal as used herein is a mammal of an age where both cellular and humoral immune responses are elicited after exposure to an antigenic stimulus, by upregulation of Th1 functions or IFN-γ production in response to polyclonal stimuli, in contrast to a neonate which has innate immunity and immunity derived from the mother, e.g., during gestation or via lactation. An adult mammal that does not have an immunodeficiency disease is an example of an immunocompetent mammal. For example, an immunocompetent human is typically at least 1, 2, 3, 4, 5 or 6 months of age, and includes adult humans without an immunodeficiency disease. In one embodiment, the AAV is administered intrathecally. In one embodiment, the MV is administered intracranially (e.g., intracerebroventricularly). In one embodiment, the AAV is administered intranasally, with or without a permeation enhancer. In one embodiment, the AAV is administered endovascularly, e.g., carotid artery administration, with or without a permeation enhancer. In one embodiment, the mammal that is administered the AAV is immunodeficient or is subjected to immunotolerization or immune suppression, e.g., to induce higher levels of therapeutic protein expression relative to a corresponding mammal that is administered the MV but not subjected to immunotolerization or immune suppression. In one embodiment, an immune suppressive agent is administered to induce immune suppression. In one embodiment, the mammal that is administered the AAV is not subjected to immunotolerization or immune suppression (e.g., administration of the AAV alone provides for the therapeutic effect).
  • The invention provides a method to prevent, inhibit or treat one or more symptoms associated with a disease or disorder of the central nervous system in a mammal in need thereof. The method includes intrathecally, e.g., to the lumbar region, or intracerebroventricularly, e.g., to the lateral ventricle, administering to the mammal a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms. In one embodiment, the gene product is a lysosomal storage enzyme. In one embodiment, the mammal is an immunocompetent adult. In one embodiment, the rAAV vector is an AAV-1, MV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, MV rh10, or AAV-9 vector. In one embodiment, the mammal is a human. In one embodiment, multiple doses are administered. In one embodiment, the composition is administered weekly, monthly or two or more months apart.
  • In one embodiment, the method includes intrathecally, e.g., to the lumbar region, administering to a mammal a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms, and optionally administering a permeation enhancer. In one embodiment, the permeation enhancer is administered before the composition. In one embodiment, the composition comprises a permeation enhancer. In one embodiment, the permeation enhancer is administered after the composition. In one embodiment, the gene product is a lysosomal storage enzyme. In one embodiment, the mammal is an immunocompetent adult. In one embodiment, the rAAV vector is an AAV-1, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV rh10, or AAV-9 vector. In one embodiment, the mammal is a human. In one embodiment, multiple doses are administered. In one embodiment, the composition is administered weekly, monthly or two or more months apart. In one embodiment, the mammal that is intrathecally administered the AAV is not subjected to immunotolerization or immune suppression (e.g., administration of the AAV alone provides for the therapeutic effect). In one embodiment, the mammal that is intrathecally administered the AAV is immunodeficient or is subjected to immunotolerization or immune suppression, e.g., to induce higher levels of therapeutic protein expression relative to a corresponding mammal that is intrathecally administered the AAV but not subjected to immunotolerization or immune suppression.
  • In one embodiment, the method includes intracerebroventricularly, e.g., to the lateral ventricle, administering to an immunocompetent mammal a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms. In one embodiment, the gene product is a lysosomal storage enzyme. In one embodiment, the rAAV vector is an AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV rh10, or AAV-9 vector. In one embodiment, the rAAV vector is not a rAAV-5 vector. In one embodiment, the mammal is a human. In one embodiment, multiple doses are administered. In one embodiment, the composition is administered weekly, monthly or two or more months apart. In one embodiment, the mammal that is intracerebroventricularly administered the AAV is not subjected to immunotolerization or immune suppression (e.g., administration of the AAV alone provides for the therapeutic effect). In one embodiment, the mammal that is intracerebroventricularly administered the AAV is immunodeficient or is subjected to immunotolerization or immune suppression, e.g., to induce higher levels of therapeutic protein expression relative to a corresponding mammal that is intracerebroventricularly administered the AAV but not subjected to immunotolerization or immune suppression In one embodiment, the mammal is immunotolerized to the gene product before the composition comprising the AAV is administered.
  • Further provided is a method to prevent, inhibit or treat one or more symptoms associated with a disease or disorder of the central nervous system in a mammal in need thereof. The method includes endovascularly administering to the mammal a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms, and an effective amount of a permeation enhancer. In one embodiment, the composition comprises the permeation enhancer. In one embodiment, the permeation enhancer comprises mannitol, sodium glycocholate, sodium taurocholate, sodium deoxycholate, sodium salicylate, sodium caprylate, sodium caprate, sodium lauryl sulfate, polyoxyethylene-9-laurel ether, or EDTA. In one embodiment, the gene product is a lysosomal storage enzyme. In one embodiment, the mammal is an immunocompetent adult. In one embodiment, the rAAV vector is an AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, MV rh10, or AAV-9 vector. In one embodiment, the rAAV vector is not a rAAV-5 vector. In one embodiment, the mammal is a human. In one embodiment, multiple doses are administered. In one embodiment, the composition is administered weekly. In one embodiment, the composition is administered weekly, monthly or two or more months apart. In one embodiment, the mammal that is endovascularly administered the AAV is not subjected to immunotolerization or immune suppression (e.g., administration of the MV provides for the therapeutic effect). In one embodiment, the mammal that is endovascularly administered the AAV is immunodeficient or is subjected to immunotolerization or immune suppression, e.g., to induce higher levels of therapeutic protein expression relative to a corresponding mammal that is endovascularly administered the AAV but not subjected to immunotolerization or immune suppression.
  • In one embodiment, the method includes intranasally administering to a mammal a composition comprising an effective amount of a rAAV-9 vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms, and optionally administering a permeation enhancer. In one embodiment, intranasal delivery may be accomplished as described in U.S. Pat. No. 8,609,088, the disclosure of which is incorporated by reference herein. In one embodiment, the permeation enhancer is administered before the composition. In one embodiment, the composition comprises a permeation enhancer. In one embodiment, the permeation enhancer is administered after the composition. In one embodiment, the gene product is a lysosomal storage enzyme. In one embodiment, the mammal is an immunocompetent adult. In one embodiment, the mammal is a human. In one embodiment, multiple doses are administered. In one embodiment, the composition is administered weekly, monthly or two or more months apart. In one embodiment, the mammal that is intranasally administered the AAV is not subjected to immunotolerization or immune suppression. In one embodiment, the mammal that is intranasally administered the AAV is subjected to immunotolerization or immune suppression, e.g., to induce higher levels of IDUA protein expression relative to a corresponding mammal that is intranasally administered the AAV but not subjected to immunotolerization or immune suppression.
  • Also provided is a method to prevent, inhibit or treat one or more symptoms associated with a disease of the central nervous system in a mammal in need thereof. The method includes administering to the mammal a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms, and an immune suppressant. In one embodiment, the immune suppressant comprises cyclophosphamide. In one embodiment, the immune suppressant comprises a glucocorticoid, cytostatic agents including an alkylating agent or an anti-metabolite such as methotrexate, azathioprine, mercaptopurine or a cytotoxic antibiotic, an antibody, or an agent active on immunophilin. In one embodiment, the immune suppressant comprises a nitrogen mustard, nitrosourea, a platinum compound, methotrexate, azathioprine, mercaptopurine, fluorouracil, dactinomycin, an anthracyclin, mitomycin C, bleomycin, mithramycin, IL2-receptor- (CD25-) or CD3-directed antibodies, anti-IL-2 antibodies, cyclosporin, tacrolimus, sirolimus, IFN-β, IFN-γ, an opioid, or TNF-α (tumor necrosis factor-alpha) binding agents such as infliximab (Remicade), etanercept (Enbrel), or adalimumab (Humira). In one embodiment, the rAAV and the immune suppressant are co-administered. In one embodiment, the rAAV is administered before and optionally after the immune suppressant. In one embodiment, the immune suppressant is administered before the rAAV. In one embodiment, the rAAV and the immune suppressant are intrathecally administered. In one embodiment, the rAAV and the immune suppressant are intracerebroventricularly administered. In one embodiment, the rAAV is intrathecally administered and the immune suppressant is intravenously administered. In one embodiment, the gene product is a lysosomal storage enzyme. In one embodiment, the mammal is an adult. In one embodiment, the rAAV vector is an AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV rh10, or AAV-9 vector. In one embodiment, the mammal is a human. In one embodiment, multiple doses are administered. In one embodiment, the composition is administered weekly. In one embodiment, the composition is administered weekly, monthly or two or more months apart.
  • The invention also provides a method to prevent, inhibit or treat one or more symptoms associated with a disease of the central nervous system in a mammal in need thereof. A mammal immunotolerized to a gene product that is associated with the disease is administered a composition comprising an effective amount of a rAAV vector comprising an open reading frame encoding a gene product, the expression of which in the central nervous system of the mammal prevents, inhibits or treats the one or more symptoms. In one embodiment, the gene product is a lysosomal storage enzyme. In one embodiment, the mammal is an adult. In one embodiment, the rAAV vector is an AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, MV rh10, or AAV-9 vector. In one embodiment, the mammal is a human. In one embodiment, multiple doses are administered. In one embodiment, the composition is administered weekly.
  • Gene products that may be encoded by rAAV vectors include, but are not limited to, alpha-L-iduronidase, iduronate-2-sulfatase, heparan sulfate sulfatase, N-acetyl-alpha-D-glucosaminidase, beta-hexosaminidase, alpha-galactosidase, betagalactosidase, beta-glucuronidase, glucocerebrosidase, fibroblast growth factor-2 (FGF-2), brain derived growth factor (BDGF), neurturin, glial derived growth factor (GDGF), tyrosine hydroxylase, dopamine decarboxylase, or glutamic acid decarboxylase.
  • Diseases that have one or more neurologic symptoms that may be prevented, inhibited or treated using the methods disclosed herein include, but are not limited to, Adrenoleukodystrophy, Alzheimer disease, Amyotrophic lateral sclerosis, Angelman syndrome, Ataxia telangiectasia, Charcot-Marie-Tooth syndrome, Cockayne syndrome, Deafness, Duchenne muscular dystrophy, Epilepsy, Essential tremor, Fragile X syndrome, Friedreich's ataxia, Gaucher disease, Huntington disease, Lesch-Nyhan syndrome, Maple syrup urine disease, Menkes syndrome, Myotonic dystrophy, Narcolepsy, Neurofibromatosis, Niemann-Pick disease, Parkinson disease, Phenylketonuria, Prader-Willi syndrome, Refsum disease, Rett syndrome, Spinal muscular atrophy, Spinocerebellar ataxia, Tangier disease, Tay-Sachs disease, Tuberous sclerosis, Von Hippel-Lindau syndrome, Williams syndrome, Wilson's disease, or Zellweger syndrome. In one embodiment, the disease is a lysosomal storage disease, e.g., a lack or deficiency in a lysosomal storage enzyme. Lysosomal storage diseases include, but are not limited to, mucopolysaccharidosis (MPS) diseases, for instance, mucopolysaccharidosis type I, e.g., Hurler syndrome and the variants Scheie syndrome and Hurler-Scheie syndrome (a deficiency in alpha-L-iduronidase); Hunter syndrome (a deficiency of iduronate-2-sulfatase); mucopolysaccharidosis type III, e.g., Sanfilippo syndrome (A, B, C or D; a deficiency of heparan sulfate sulfatase, N-acetyl-alpha-D-glucosaminidase, acetyl CoA:alpha-glucosaminide N-acetyl transferase or N-acetylglucosamine-6-sulfate sulfatase); mucopolysaccharidosis type IV e.g., mucopolysaccharidosis type IV, e.g., Morquio syndrome (a deficiency of galactosamine-6-sulfate sulfatase or beta-galactosidase); mucopolysaccharidosis type VI, e.g., Maroteaux-Lamy syndrome (a deficiency of arylsulfatase B); mucopolysaccharidosis type II; mucopolysaccharidosis type III (A, B, C or D; a deficiency of heparan sulfate sulfatase, N-acetyl-alpha-D-glucosaminidase, acetyl CoA:alpha-glucosaminide N-acetyl transferase or N-acetylglucosamine-6-sulfate sulfatase); mucopolysaccharidosis type IV (A or B; a deficiency of galactosamine-6-sulfatase and beta-galatacosidase); mucopolysaccharidosis type VI (a deficiency of arylsulfatase B); mucopolysaccharidosis type VII (a deficiency in beta-glucuronidase); mucopolysaccharidosis type VIII (a deficiency of glucosamine-6-sulfate sulfatase); mucopolysaccharidosis type IX (a deficiency of hyaluronidase); Tay-Sachs disease (a deficiency in alpha subunit of beta-hexosaminidase); Sandhoff disease (a deficiency in both alpha and beta subunit of beta-hexosaminidase); GM1 gangliosidosis (type I or type II); Fabry disease (a deficiency in alpha galactosidase); metachromatic leukodystrophy (a deficiency of aryl sulfatase A); Pompe disease (a deficiency of acid maltase); fucosidosis (a deficiency of fucosidase); alpha-mannosidosis (a deficiency of alpha-mannosidase); beta-mannosidosis (a deficiency of beta-mannosidase), ceroid lipofuscinosis, and Gaucher disease (types I, II and III; a deficiency in glucocerebrosidase), as well as disorders such as Hermansky-Pudlak syndrome; Amaurotic idiocy; Tangier disease; aspartylglucosaminuria; congenital disorder of glycosylation, type Ia; Chediak-Higashi syndrome; macular dystrophy, corneal, 1; cystinosis, nephropathic; Fanconi-Bickel syndrome; Farber lipogranulomatosis; fibromatosis; geleophysic dysplasia; glycogen storage disease I; glycogen storage disease Ib; glycogen storage disease Ic; glycogen storage disease III; glycogen storage disease IV; glycogen storage disease V; glycogen storage disease VI; glycogen storage disease VII; glycogen storage disease 0; immunoosseous dysplasia, Schimke type; lipidosis; lipase b; mucolipidosis II; mucolipidosis II, including the variant form; mucolipidosis IV; neuraminidase deficiency with beta-galactosidase deficiency; mucolipidosis I; Niemann-Pick disease (a deficiency of sphingomyelinase); Niemann-Pick disease without sphingomyelinase deficiency (a deficiency of a npc1 gene encoding a cholesterol metabolizing enzyme); Refsum disease; Sea-blue histiocyte disease; infantile sialic acid storage disorder; sialuria; multiple sulfatase deficiency; triglyceride storage disease with impaired long-chain fatty acid oxidation; Winchester disease; Wolman disease (a deficiency of cholesterol ester hydrolase); Deoxyribonuclease I-like 1 disorder; arylsulfatase E disorder; ATPase, H+ transporting, lysosomal, subunit 1 disorder; glycogen storage disease Ilb; Ras-associated protein rab9 disorder; chondrodysplasia punctata 1, X-linked recessive disorder; glycogen storage disease VIII; lysosome-associated membrane protein 2 disorder; Menkes syndrome; congenital disorder of glycosylation, type Ic; and sialuria. Replacement of less than 20%, e.g., less than 10% or about 1% to 5% levels of lysosomal storage enzyme found in nondiseased mammals, may prevent, inhibit or treat neurological symptoms such as neurological degeneration in mammals.
  • In one embodiment, the methods described herein involve delivering to the CNS of an immunocompetent human in need of treatment a composition comprising an effective amount of a rAAV-9 vector comprising an open reading frame encoding a IDUA. Routes of administration to the CNS/brain include, but are not limited to intrathecal administration, intracranial administration, e.g., intracerebroventricular administration or lateral cerebro ventricular administration, intranasal administration, endovascular administration, and intraparenchymal administration.
  • Other viral vectors may be employed in the methods of the invention, e.g., viral vectors such as retrovirus, lentivirus, adenovirus, semliki forest virus or herpes simplex virus vectors.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1. Experimental design for iduronidase-deficient adult mice administered IDUA-AAV either intracerebroventricularly (ICV) (right lateral ventricle) or intrathecally (lumbar area). To prevent immune response, animals were either immunosuppressed with cyclophosphamide (CP), immunotolerized at birth by intravenous administration of human iduonidase protein (aldurazyme), or the injections (10 microliters, 3×1011 particles) were carried out in NOD-SCID immunodeficient mice that were also iduronidase deficient. Animals were sacrificed at the indicated time post-treatment, the brains were microdissected and extracts assayed for iduronidase activity.
  • FIGS. 2A-B. IDUA activity in immunodeficient, IDUA deficient animals after ICV injection.
  • FIGS. 3A-B. IDUA activity in immunocompetent animals administered AAV vector by ICV route and treated with cyclophosphamide.
  • FIGS. 4A-B. IDUA activity in immunocompetent animals administered AAV vector by IT route and treated with cyclophosphamide.
  • FIGS. 5A-B. IDUA activity in immunotolerized immunocompetent animals administered AAV vector ICV.
  • FIG. 6. Compilation of all mean levels of IDUA activity for side-by-side comparison.
  • FIGS. 7A-E. Data are grouped according the area of the brain. A) hippocampus, B) cerebellum, C) cortex, D) striatum, and E) rest of brain.
  • FIGS. 8A-D. Assay for GAG storage material in the different sections of the brain for all four of the test groups. A) Immunodeficient mice, ICV administration. B) Immunocompetent mice, ICV administration and CP administration. C) Immunocompetent mice, ICV administration and aldurazyme administration. D) Immunocompetent mice, IT administration and CP administration.
  • FIG. 9. Schematic of experimental design. All mice were adult MPS1 (IDUA deficient) mice injected with 10 microliters (3×1011 particles) of AAV.
  • FIGS. 10A-B. Intracranial infusion of AAV9IDUA into immunodeficient MPS I mice. Adult animals were injected with 1011 vector genomes and evaluated for iduronidase expression in the brain after 10 weeks. Enzyme activity levels in the brain were significantly higher than in the brains of wild type animals, and ranged from 30- to 300-fold higher than wild type.
  • FIGS. 11A-B. Intracranial administration of AAV9IDUA in immunocompetent, IDUA deficient mice. Adult animals were injected with 1011 vector genomes, and immunosuppressed by weekly injection of cyclophosphamide (CP). CP injections were terminated at 6 weeks post vector injection due to poor health, and the animals were sacrificed at 8 weeks post-injection. Brains were microdissected and assayed for IDUA enzyme activity.
  • FIGS. 12A-B. Intracranial infusion of AAV9IDUA into immunotolerized MPS I mice. MPS 1 mice were tolerized with either a single dose of Aldurazyme at birth or multiple doses administered weekly, starting at birth. Mice were infused with vector at 4 months, and sacrificed at 11 weeks after injection. Brains were microdissected and analyzed for iduronidase expression. Enzyme activities ranged from an average of 10- to 1000-fold higher than wild type levels.
  • FIGS. 13A-B. Intrathecal administration of AAV9IDUA in immunocompetent, IDUA deficient animals. Adult MPS I mice were injected with AAV9IDUA intrathecally, followed by a weekly immunosuppressive regimen of cyclophosphamide. Animals were sacrificed at 11 weeks post-injection, and then brains and spinal cords were analyzed for IDUA enzyme activity.
  • FIGS. 14A-B. Intrathecal infusion of AAV9IDUA in immunotolerized MPS I mice. IDUA deficient animals were tolerized at birth with a single dose of Aldurazyme or multiple doses administered weekly starting at birth. At 4 months of age animals were infused intrathecally with AAV9IDUA vector, and at 10 weeks post-injection animals were sacrificed, brains microdissected and assayed for iduronidase activity. There was restoration of enzyme activity in all parts of the brain, with activities in the cerebellum ranging from 200- to 1500-fold higher than wild type levels. Levels of enzyme activity in the olfactory bulb and cerebellum (to the right of the dashed line) correspond to the right Y-axis.
  • FIGS. 15A-B. Intrathecal infusion of AAV9IDUA in immunocompetent MPS I animals. Control MPS I animals were injected with AAV9IDUA vector, but were not immunosuppressed nor immunotolerized. Animals were sacrificed at 11 weeks after vector injection, and then their brains were assayed for iduronidase activity. Enzyme levels were restored to wild type levels in all parts of the brain, but were significantly lower than in animals that were either immunosuppressed or immunotolerized.
  • FIG. 16. Normalization of glycosaminoglycan (GAG) levels following intracranial or intrathecal AAV9 infusion. AAV9IDUA was injected intracranially or intrathecally into immunodeficient, immunosuppressed or immunotolerized MPS I mice as indicated. Animals were sacrificed 8-11 weeks after injection, then the brains were microdissected and analyzed for GAG levels. GAG storage was restored to wild type levels or close to wild type in all groups analyzed. H=hippocampus, Cb=cerebellum, C=cortex, S=striatum, ThB=thalamus and brain stem.
  • FIGS. 17A-B. IDUA vector copies in brain. Microdissected brains were analyzed for IDUA vector sequences by QPCR. The copy numbers in intracranially (A) and intrathecally (B) injected mice correlate to the levels of enzyme activity depicted in FIGS. 11 and 13.
  • FIG. 18. ICV infusion of AAV8-MCI into adult animals.
  • FIGS. 19A-B. Intranasal administration of AAV9/IDUA in immunocompetent, IDUA deficient animals. Adult MPS I mice were infused with AAV9/IDUA intranasally, followed by a weekly immunosuppressive regimen of cyclophosphamide. Animals were sacrificed at 12 weeks post-injection and brains were analyzed for IDUA enzyme activity.
  • FIG. 20. IDUA vector copies in brain. Microdissected brains were analyzed for IDUA vector sequences by QPCR. The copy numbers in intranasally injected mice correlate to the levels of enzyme in FIG. 19.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • As used herein, “individual” (as in the subject of the treatment) means a mammal. Mammals include, for example, humans; non-human primates, e.g., apes and monkeys; and non-primates, e.g., dogs, cats, rats, mice, cattle, horses, sheep, and goats. Non-mammals include, for example, fish and birds.
  • The term “disease” or “disorder” are used interchangeably, and are used to refer to diseases or conditions wherein lack of or reduced amounts of a specific gene product, e.g., a lysosomal storage enzyme, plays a role in the disease such that a therapeutically beneficial effect can be achieved by supplementing, e.g., to at least 1% of normal levels.
  • “Substantially” as the term is used herein means completely or almost completely; for example, a composition that is “substantially free” of a component either has none of the component or contains such a trace amount that any relevant functional property of the composition is unaffected by the presence of the trace amount, or a compound is “substantially pure” is there are only negligible traces of impurities present.
  • “Treating” or “treatment” within the meaning herein refers to an alleviation of symptoms associated with a disorder or disease, “inhibiting” means inhibition of further progression or worsening of the symptoms associated with the disorder or disease, and “preventing” refers to prevention of the symptoms associated with the disorder or disease.
  • As used herein, an “effective amount” or a “therapeutically effective amount” of an agent of the invention e.g., a recombinant AAV encoding a gene product, refers to an amount of the agent that alleviates, in whole or in part, symptoms associated with the disorder or condition, or halts or slows further progression or worsening of those symptoms, or prevents or provides prophylaxis for the disorder or condition, e.g., an amount that is effective to prevent, inhibit or treat in the individual one or more neurological symptoms.
  • In particular, a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount is also one in which any toxic or detrimental effects of compounds of the invention are outweighed by the therapeutically beneficial effects.
  • A “vector” as used herein refers to a macromolecule or association of macromolecules that comprises or associates with a polynucleotide and which can be used to mediate delivery of the polynucleotide to a cell, either in vitro or in vivo. Illustrative vectors include, for example, plasmids, viral vectors, liposomes and other gene delivery vehicles. The polynucleotide to be delivered, sometimes referred to as a “target polynucleotide” or “transgene,” may comprise a coding sequence of interest in gene therapy (such as a gene encoding a protein of therapeutic interest) and/or a selectable or detectable marker.
  • “AAV” is adeno-associated virus, and may be used to refer to the virus itself or derivatives thereof. The term covers all subtypes, serotypes and pseudotypes, and both naturally occurring and recombinant forms, except where required otherwise. As used herein, the term “serotype” refers to an AAV which is identified by and distinguished from other AAVs based on its binding properties, e.g., there are eleven serotypes of AAVs, AAV-1-AAV-11, including AAV-2, AAV-5, AAV-8, AAV-9 and AAV rh10, and the term encompasses pseudotypes with the same binding properties. Thus, for example, AAV-5 serotypes include AAV with the binding properties of AAV-5, e.g., a pseudotyped AAV comprising AAV-5 capsid and a rAAV genome which is not derived or obtained from AAV-5 or which genome is chimeric. The abbreviation “rAAV” refers to recombinant adeno-associated virus, also referred to as a recombinant AAV vector (or “rAAV vector”).
  • An “AAV virus” refers to a viral particle composed of at least one AAV capsid protein and an encapsidated polynucleotide. If the particle comprises a heterologous polynucleotide (i.e., a polynucleotide other than a wild-type AAV genome such as a transgene to be delivered to a mammalian cell), it is typically referred to as “rAAV”. An MV “capsid protein” includes a capsid protein of a wild-type AAV, as well as modified forms of an AAV capsid protein which are structurally and or functionally capable of packaging a rAAV genome and bind to at least one specific cellular receptor which may be different than a receptor employed by wild type AAV. A modified AAV capsid protein includes a chimeric AAV capsid protein such as one having amino acid sequences from two or more serotypes of AAV, e.g., a capsid protein formed from a portion of the capsid protein from AAV-5 fused or linked to a portion of the capsid protein from MV-2, and a AAV capsid protein having a tag or other detectable non-AAV capsid peptide or protein fused or linked to the AAV capsid protein, e.g., a portion of an antibody molecule which binds the transferrin receptor may be recombinantly fused to the AAV-2 capsid protein.
  • A “pseudotyped” rAAV is an infectious virus having any combination of an AAV capsid protein and an AAV genome. Capsid proteins from any AAV serotype may be employed with a rAAV genome which is derived or obtainable from a wild-type AAV genome of a different serotype or which is a chimeric genome, i.e., formed from AAV DNA from two or more different serotypes, e.g., a chimeric genome having 2 inverted terminal repeats (ITRs), each ITR from a different serotype or chimeric ITRs. The use of chimeric genomes such as those comprising ITRs from two AAV serotypes or chimeric ITRs can result in directional recombination which may further enhance the production of transcriptionally active intermolecular concatamers. Thus, the 5′ and 3′ ITRs within a rAAV vector of the invention may be homologous, i.e., from the same serotype, heterologous, i.e., from different serotypes, or chimeric, i.e., an ITR which has ITR sequences from more than one AAV serotype.
  • rAAV Vectors
  • Adeno-associated viruses of any serotype are suitable to prepare rAAV, since the various serotypes are functionally and structurally related, even at the genetic level. All AAV serotypes apparently exhibit similar replication properties mediated by homologous rep genes; and all generally bear three related capsid proteins such as those expressed in AAV2. The degree of relatedness is further suggested by heteroduplex analysis which reveals extensive cross-hybridization between serotypes along the length of the genome; and the presence of analogous self-annealing segments at the termini that correspond to ITRs. The similar infectivity patterns also suggest that the replication functions in each serotype are under similar regulatory control. Among the various AAV serotypes, AAV2 is most commonly employed.
  • An AAV vector of the invention typically comprises a polynucleotide that is heterologous to AAV. The polynucleotide is typically of interest because of a capacity to provide a function to a target cell in the context of gene therapy, such as up- or down-regulation of the expression of a certain phenotype. Such a heterologous polynucleotide or “transgene,” generally is of sufficient length to provide the desired function or encoding sequence.
  • Where transcription of the heterologous polynucleotide is desired in the intended target cell, it can be operably linked to its own or to a heterologous promoter, depending for example on the desired level and/or specificity of transcription within the target cell, as is known in the art. Various types of promoters and enhancers are suitable for use in this context. Constitutive promoters provide an ongoing level of gene transcription, and may be preferred when it is desired that the therapeutic or prophylactic polynucleotide be expressed on an ongoing basis. Inducible promoters generally exhibit low activity in the absence of the inducer, and are up-regulated in the presence of the inducer. They may be preferred when expression is desired only at certain times or at certain locations, or when it is desirable to titrate the level of expression using an inducing agent. Promoters and enhancers may also be tissue-specific: that is, they exhibit their activity only in certain cell types, presumably due to gene regulatory elements found uniquely in those cells.
  • Illustrative examples of promoters are the SV40 late promoter from simian virus 40, the Baculovirus polyhedron enhancer/promoter element, Herpes Simplex Virus thymidine kinase (HSV tk), the immediate early promoter from cytomegalovirus (CMV) and various retroviral promoters including LTR elements. Inducible promoters include heavy metal ion inducible promoters (such as the mouse mammary tumor virus (mMTV) promoter or various growth hormone promoters), and the promoters from T7 phage which are active in the presence of T7 RNA polymerase. By way of illustration, examples of tissue-specific promoters include various surfactin promoters (for expression in the lung), myosin promoters (for expression in muscle), and albumin promoters (for expression in the liver). A large variety of other promoters are known and generally available in the art, and the sequences of many such promoters are available in sequence databases such as the GenBank database.
  • Where translation is also desired in the intended target cell, the heterologous polynucleotide will preferably also comprise control elements that facilitate translation (such as a ribosome binding site or “RBS” and a polyadenylation signal). Accordingly, the heterologous polynucleotide generally comprises at least one coding region operatively linked to a suitable promoter, and may also comprise, for example, an operatively linked enhancer, ribosome binding site and poly-A signal. The heterologous polynucleotide may comprise one encoding region, or more than one encoding regions under the control of the same or different promoters. The entire unit, containing a combination of control elements and encoding region, is often referred to as an expression cassette.
  • The heterologous polynucleotide is integrated by recombinant techniques into or in place of the AAV genomic coding region (i.e., in place of the AAV rep and cap genes), but is generally flanked on either side by AAV inverted terminal repeat (ITR) regions. This means that an ITR appears both upstream and downstream from the coding sequence, either in direct juxtaposition, e.g., (although not necessarily) without any intervening sequence of AAV origin in order to reduce the likelihood of recombination that might regenerate a replication-competent AAV genome. However, a single ITR may be sufficient to carry out the functions normally associated with configurations comprising two ITRs (see, for example, WO 94/13788), and vector constructs with only one ITR can thus be employed in conjunction with the packaging and production methods of the present invention.
  • The native promoters for rep are self-regulating, and can limit the amount of AAV particles produced. The rep gene can also be operably linked to a heterologous promoter, whether rep is provided as part of the vector construct, or separately. Any heterologous promoter that is not strongly down-regulated by rep gene expression is suitable; but inducible promoters may be preferred because constitutive expression of the rep gene can have a negative impact on the host cell. A large variety of inducible promoters are known in the art; including, by way of illustration, heavy metal ion inducible promoters (such as metallothionein promoters); steroid hormone inducible promoters (such as the MMTV promoter or growth hormone promoters); and promoters such as those from T7 phage which are active in the presence of T7 RNA polymerase. One sub-class of inducible promoters are those that are induced by the helper virus that is used to complement the replication and packaging of the rAAV vector. A number of helper-virus-inducible promoters have also been described, including the adenovirus early gene promoter which is inducible by adenovirus E1A protein; the adenovirus major late promoter; the herpesvirus promoter which is inducible by herpesvirus proteins such as VP16 or 1CP4; as well as vaccinia or poxvirus inducible promoters.
  • Methods for identifying and testing helper-virus-inducible promoters have been described (see, e.g., WO 96/17947). Thus, methods are known in the art to determine whether or not candidate promoters are helper-virus-inducible, and whether or not they will be useful in the generation of high efficiency packaging cells. Briefly, one such method involves replacing the p5 promoter of the MV rep gene with the putative helper-virus-inducible promoter (either known in the art or identified using well-known techniques such as linkage to promoter-less “reporter” genes). The MV rep-cap genes (with p5 replaced), e.g., linked to a positive selectable marker such as an antibiotic resistance gene, are then stably integrated into a suitable host cell (such as the HeLa or A549 cells exemplified below). Cells that are able to grow relatively well under selection conditions (e.g., in the presence of the antibiotic) are then tested for their ability to express the rep and cap genes upon addition of a helper virus. As an initial test for rep and/or cap expression, cells can be readily screened using immunofluorescence to detect Rep and/or Cap proteins. Confirmation of packaging capabilities and efficiencies can then be determined by functional tests for replication and packaging of incoming rAAV vectors. Using this methodology, a helper-virus-inducible promoter derived from the mouse metallothionein gene has been identified as a suitable replacement for the p5 promoter, and used for producing high titers of rAAV particles (as described in WO 96/17947).
  • Removal of one or more AAV genes is in any case desirable, to reduce the likelihood of generating replication-competent AAV (“RCA”). Accordingly, encoding or promoter sequences for rep, cap, or both, may be removed, since the functions provided by these genes can be provided in trans.
  • The resultant vector is referred to as being “defective” in these functions. In order to replicate and package the vector, the missing functions are complemented with a packaging gene, or a plurality thereof, which together encode the necessary functions for the various missing rep and/or cap gene products. The packaging genes or gene cassettes are in one embodiment not flanked by AAV ITRs and in one embodiment do not share any substantial homology with the rAAV genome. Thus, in order to minimize homologous recombination during replication between the vector sequence and separately provided packaging genes, it is desirable to avoid overlap of the two polynucleotide sequences. The level of homology and corresponding frequency of recombination increase with increasing length of homologous sequences and with their level of shared identity. The level of homology that will pose a concern in a given system can be determined theoretically and confirmed experimentally, as is known in the art. Typically, however, recombination can be substantially reduced or eliminated if the overlapping sequence is less than about a 25 nucleotide sequence if it is at least 80% identical over its entire length, or less than about a 50 nucleotide sequence if it is at least 70% identical over its entire length. Of course, even lower levels of homology are preferable since they will further reduce the likelihood of recombination. It appears that, even without any overlapping homology, there is some residual frequency of generating RCA. Even further reductions in the frequency of generating RCA (e.g., by nonhomologous recombination) can be obtained by “splitting” the replication and encapsidation functions of AAV, as described by Allen et al., WO 98/27204).
  • The rAAV vector construct, and the complementary packaging gene constructs can be implemented in this invention in a number of different forms. Viral particles, plasmids, and stably transformed host cells can all be used to introduce such constructs into the packaging cell, either transiently or stably.
  • In certain embodiments of this invention, the AAV vector and complementary packaging gene(s), if any, are provided in the form of bacterial plasmids, AAV particles, or any combination thereof. In other embodiments, either the AAV vector sequence, the packaging gene(s), or both, are provided in the form of genetically altered (preferably inheritably altered) eukaryotic cells. The development of host cells inheritably altered to express the AAV vector sequence, AAV packaging genes, or both, provides an established source of the material that is expressed at a reliable level.
  • A variety of different genetically altered cells can thus be used in the context of this invention. By way of illustration, a mammalian host cell may be used with at least one intact copy of a stably integrated rAAV vector. An AAV packaging plasmid comprising at least an AAV rep gene operably linked to a promoter can be used to supply replication functions (as described in U.S. Pat. No. 5,658,776). Alternatively, a stable mammalian cell line with an AAV rep gene operably linked to a promoter can be used to supply replication functions (see, e.g., Trempe et al., WO 95/13392); Burstein et al. (WO 98/23018); and Johnson et al. (U.S. Pat. No. 5,656,785). The AAV cap gene, providing the encapsidation proteins as described above, can be provided together with an AAV rep gene or separately (see, e.g., the above-referenced applications and patents as well as Allen et al. (WO 98/27204). Other combinations are possible and included within the scope of this invention.
  • Pathways for Delivery
  • Despite the immense network of the cerebral vasculature, systemic delivery of therapeutics to the central nervous system (CNS) is not effective for greater than 98% of small molecules and for nearly 100% of large molecules (Partridge, 2005). The lack of effectiveness is due to the presence of the blood-brain barrier (BBB), which prevents most foreign substances, even many beneficial therapeutics, from entering the brain from the circulating blood. While certain small molecule, peptide, and protein therapeutics given systemically reach the brain parenchyma by crossing the BBB (Banks, 2008), generally high systemic doses are needed to achieve therapeutic levels, which can lead to adverse effects in the body. Therapeutics can be introduced directly into the CNS by intracerebroventricular or intraparenchymal injections. Intranasal delivery bypasses the BBB and targets therapeutics directly to the CNS utilizing pathways along olfactory and trigeminal nerves innervating the nasal passages (Frey II, 2002; Thorne et al., 2004; Dhanda et al., 2005).
  • Any route of rAAV administration may be employed so long as that route and the amount administered are prophylactically or therapeutically useful. In one example, routes of administration to the CNS include intrathecal and intracranial. Intracranial administration may be to the cisterna magna or ventricle. The term “cisterna magna” is intended to include access to the space around and below the cerebellum via the opening between the skull and the top of the spine. The term “cerebral ventricle” is intended to include the cavities in the brain that are continuous with the central canal of the spinal cord. Intracranial administration is via injection or infusion and suitable dose ranges for intracranial administration are generally about 103 to 1015 infectious units of viral vector per microliter delivered in 1 to 3000 microliters of single injection volume. For instance, viral genomes or infectious units of vector per micro liter would generally contain about 104, 105, 106, 107, 108, 109, 1010, 1011, 1012, 1013, or 1014 viral genomes or infectious units of viral vector delivered in about 10, 50, 100, 200, 500, 1000, or 2000 microliters. It should be understood that the aforementioned dosage is merely an exemplary dosage and those of skill in the art will understand that this dosage may be varied. Effective doses may be extrapolated from dose-responsive curves derived from in vitro or in vivo test systems.
  • The AAV delivered in the intrathecal methods of treatment of the present invention may be administered through any convenient route commonly used for intrathecal administration. For example, the intrathecal administration may be via a slow infusion of the formulation for about an hour. Intrathecal administration is via injection or infusion and suitable dose ranges for intrathecal administration are generally about 103 to 1015 infectious units of viral vector per microliter delivered in, for example, 1 to 3000 microliters or 0.5 to 15 milliliters of single injection volume. For instance, viral genomes or infectious units of vector per microliter would generally contain about 104, 105, 106, 107, 108, 109, 1010, 1011, 1012, 1013, or 1014 viral genomes or infectious units of viral vector.
  • The therapy, if a lysosomal storage enzyme such as IDUA is expressed, results in the normalization of lysosomal storage granules in the neuronal and/or meningeal tissue of the subjects as discussed above. It is contemplated that the deposition of storage granules is ameliorated from neuronal and glial tissue, thereby alleviating the developmental delay and regression seen in individuals suffering with lysosomal storage disease. Other effects of the therapy may include the normalization of lysosomal storage granules in the cerebral meninges near the arachnoid granulation, the presence of which in lysosomal storage disease result in high pressure hydrocephalus. The methods of the invention also may be used in treating spinal cord compression that results from the presence of lysosomal storage granules in the cervical meninges near the cord at C1-05 or elsewhere in the spinal cord. The methods of the invention also are directed to the treatment of cysts that are caused by the perivascular storage of lysosomal storage granules around the vessels of the brain. In other embodiments, the therapy also may advantageously result in normalization of liver volume and urinary glycosaminoglycan excretion, reduction in spleen size and apnea/hypopnea events, increase in height and growth velocity in prepubertal subjects, increase in shoulder flexion and elbow and knee extension, and reduction in tricuspid regurgitation or pulmonic regurgitation.
  • The intrathecal administration of the present invention may comprise introducing the composition into the lumbar area. Any such administration may be via a bolus injection. Depending on the severity of the symptoms and the responsiveness of the subject to the therapy, the bolus injection may be administered once per week, once per month, once every 6 months or annually. In other embodiments, the intrathecal administration is achieved by use of an infusion pump. Those of skill in the art are aware of devices that may be used to effect intrathecal administration of a composition. The composition may be intrathecally given, for example, by a single injection, or continuous infusion. It should be understood that the dosage treatment may be in the form of a single dose administration or multiple doses.
  • As used herein, the term “intrathecal administration” is intended to include delivering a pharmaceutical composition directly into the cerebrospinal fluid of a subject, by techniques including lateral cerebroventricular injection through a burrhole or cistemal or lumbar puncture or the like. The term “lumbar region” is intended to include the area between the third and fourth lumbar (lower back) vertebrae and, more inclusively, the L2-S1 region of the spine.
  • Administration of a composition in accordance with the present invention to any of the above mentioned sites can be achieved by direct injection of the composition or by the use of infusion pumps. For injection, the composition can be formulated in liquid solutions, e.g., y in physiologically compatible buffers such as Hank's solution, Ringer's solution or phosphate buffer. In addition, the enzyme may be formulated in solid form and re-dissolved or suspended immediately prior to use. Lyophilized forms are also included. The injection can be, for example, in the form of a bolus injection or continuous infusion (e.g., using infusion pumps) of the enzyme.
  • In one embodiment of the invention, the rAAV is administered by lateral cerebro-ventricular injection into the brain of a subject. The injection can be made, for example, through a burr hole made in the subject's skull. In another embodiment, the enzyme and/or other pharmaceutical formulation is administered through a surgically inserted shunt into the cerebral ventricle of a subject. For example, the injection can be made into the lateral ventricles, which are larger, even though injection into the third and fourth smaller ventricles can also be made. In yet another embodiment, the compositions used in the present invention are administered by injection into the cistema magna or lumbar area of a subject.
  • While the exact mechanisms underlying intranasal drug delivery to the CNS are not entirely understood, an accumulating body of evidence demonstrates that pathways involving nerves connecting the nasal passages to the brain and spinal cord are important. In addition, pathways involving the vasculature, cerebrospinal fluid, and lymphatic system have been implicated in the transport of molecules from the nasal cavity to the CNS. It is likely that a combination of these pathways is responsible, although one pathway may predominate, depending on the properties of the therapeutic, the characteristics of the formulation, and the delivery device used.
  • Therapeutics can rapidly gain access to the CNS following intranasal administration along olfactory nerve pathways leading from the nasal cavity directly to the CNS. Olfactory nerve pathways are a major component of intranasal delivery, evidenced by the fact that fluorescent tracers are associated with olfactory nerves as they traverse the cribriform plate (Jansson et al., 2002), drug concentrations in the olfactory bulbs are generally among the highest CNS concentrations observed (Thorne et al., 2004; Banks et al., 2004; Graff et al., 2005a); Nonaka et al., 2008; Ross et al., 2004; Ross et al., 2008; Thorne et al., 2008), and a strong, positive correlation exists between concentrations in the olfactory epithelium and olfactory bulbs (Dhuria et al., 2009a).
  • Olfactory pathways arise in the upper portion of the nasal passages, in the olfactory region, where olfactory receptor neurons (ORNs) are interspersed among supporting cells (sustentacular cells), microvillar cells, and basal cells. ORNs mediate the sense of smell by conveying sensory information from the peripheral environment to the CNS (Clerico et al., 2003). Beneath the epithelium, the lamina propria contains mucus secreting Bowman's glands, axons, blood vessels, lymphatic vessels, and connective tissue. The dendrites of ORNs extend into the mucous layer of the olfactory epithelium, while axons of these bipolar neurons extend centrally through the lamina propria and through perforations in the cribriform plate of the ethmoid bone, which separates the nasal and cranial cavities. The axons of ORNs pass through the subarachnoid space containing CSF and terminate on mitral cells in the olfactory bulbs. From there, neural projections extend to multiple brain regions including the olfactory tract, anterior olfactory nucleus, piriform cortex, amygdala, and hypothalamus (Buck, 2000). In addition to ORNs, chemosensory neurons located at the anterior tip of the nasal cavity in the Grueneberg ganglion lead into the olfactory bulbs (Fuss et al., 2005; Koos et al., 2005).
  • The unique characteristics of the ORNs contribute to a dynamic cellular environment critical for intranasal delivery to the CNS. Due to the direct contact with toxins in the external environment, ORNs regenerate every 3-4 weeks from basal cells residing in the olfactory epithelium (Mackay-Sim, 2003). Special Schwann cell-like cells called olfactory ensheathing cells (OECs) envelope the axons of ORNs and have an important role in axonal regeneration, regrowth, and remyelination (Field et al., 2003; Li et al., 2005a; Li et al., 2005b). The OECs create continuous, fluid-filled perineurial channels that, interestingly, remain open, despite the degeneration and regeneration of ORNs (Williams et al., 2004).
  • Given the unique environment of the olfactory epithelium, it is possible for intranasally administered therapeutics to reach the CNS via extracellular or intracellular mechanisms of transport along olfactory nerves. Extracellular transport mechanisms involve the rapid movement of molecules between cells in the nasal epithelium, requiring only several minutes to 30 minutes for a drug to reach the olfactory bulbs and other areas of the CNS after intranasal administration (Frey II, 2002; Balin et al., 1986). Transport likely involves bulk flow mechanisms (Thorne et al., 2004; Thorne et al., 2001) within the channels created by the OECs. Drugs may also be propelled within these channels by the structural changes that occur during depolarization and axonal propagation of the action potential in adjacent axons (Luzzati et al., 2004). Intracellular transport mechanisms involve the uptake of molecules into ORNs by passive diffusion, receptor-mediated endocytosis or adsorptive endocytosis, followed by slower axonal transport, taking several hours to days for a drug to appear in the olfactory bulbs and other brain areas (Baker et al., 1986; Broadwell et al., 1985; Kristensson et al., 1971). Intracellular transport in ORNs has been demonstrated for small, lipophilic molecules such as gold particles (de Lorenzo, 1970; Gopinath et al., 1978), aluminum salts (Perl et al., 1987), and for substances with receptors on ORNs such as WGA-HRP (Thorne et al., 1995; Baker et al., 1986; Itaya et al., 1986; Shipley, 1985). Intracellular mechanisms, while important for certain therapeutics, are not likely to be the predominant mode of transport into the CNS. While some large molecules, such as galanin-like peptide (GALP), exhibit saturable transport pathways into the CNS (Nonaka et al., 2008), for other large molecules such as NGF and insulin-like growth factor-I (IGF-I), intranasal delivery into the brain is nonsaturable and not receptor mediated (Thorne et al., 2004; Chen et al., 1998; Zhao et al., 2004),
  • An often overlooked but important pathway connecting the nasal passages to the CNS involves the trigeminal nerve, which innervates the respiratory and olfactory epithelium of the nasal passages and enters the CNS in the pons (Clerico et al., 2003; Graff et al., 2003). Interestingly, a small portion of the trigeminal nerve also terminates in the olfactory bulbs (Schaefer et al., 2002). The cellular composition of the respiratory region of the nasal passages is different from that of the olfactory region, with ciliated epithelial cells distributed among mucus secreting goblet cells. These cells contribute to mucociliary clearance mechanisms that remove mucus along with foreign substances from the nasal cavity to the nasopharynx. The trigeminal nerve conveys sensory information from the nasal cavity, the oral cavity, the eyelids, and the cornea, to the CNS via the ophthalmic division (V1), the maxillary division (V2), or the mandibular division (V3) of the trigeminal nerve (Clerico et al., 2003; Gray, 1978). Branches from the ophthalmic division of the trigeminal nerve provide innervation to the dorsal nasal mucosa and the anterior portion of the nose, while branches of the maxillary division provide innervation to the lateral walls of the nasal mucosa. The mandibular division of the trigeminal nerve extends to the lower jaw and teeth, with no direct neural inputs to the nasal cavity. The three branches of the trigeminal nerve come together at the trigeminal ganglion and extend centrally to enter the brain at the level of the pons, terminating in the spinal trigeminal nuclei in the brainstem. A unique feature of the trigeminal nerve is that it enters the brain from the respiratory epithelium of the nasal passages at two sites: (1) through the anterior lacerated foramen near the pons and (2) through the cribriform plate near the olfactory bulbs, creating entry points into both caudal and rostral brain areas following intranasal administration. It is also likely that other nerves that innervate the face and head, such as the facial nerve, or other sensory structures in the nasal cavity, such as the Grueneberg ganglion, may provide entry points for intranasally applied therapeutics into the CNS.
  • Traditionally, the intranasal route of administration has been utilized to deliver drugs to the systemic circulation via absorption into the capillary blood vessels underlying the nasal mucosa. The nasal mucosa is highly vascular, receiving its blood supply from branches of the maxillary, ophthalmic and facial arteries, which arise from the carotid artery (Clerico et al., 2003; Cauna, 1982). The olfactory mucosa receives blood from small branches of the ophthalmic artery, whereas the respiratory mucosa receives blood from a large caliber arterial branch of the maxillary artery (DeSesso, 1993). The relative density of blood vessels is greater in the respiratory mucosa compared to the olfactory mucosa, making the former region an ideal site for absorption into the blood (DeSesso, 1993). The vasculature in the respiratory region contains a mix of continuous and fenestrated endothelia (Grevers et al., 1987; Van Diest et al., 1979), allowing both small and large molecules to enter the systemic circulation following nasal administration.
  • Delivery to the CNS following absorption into the systemic circulation and subsequent transport across the BBB is possible, especially for small, lipophilic drugs, which more easily enter the blood stream and cross the BBB compared to large, hydrophilic therapeutics such as peptides and proteins.
  • Increasing evidence is emerging suggesting that mechanisms involving channels associated with blood vessels, or perivascular channels, are involved in intranasal drug delivery to the CNS. Perivascular spaces are bound by the outermost layer of blood vessels and the basement membrane of the surrounding tissue (Pollock et al., 1997). These perivascular spaces act as a lymphatic system for the brain, where neuron-derived substances are cleared from brain interstitial fluid by entering perivascular channels associated with cerebral blood vessels. Perivascular transport is due to bulk flow mechanisms, as opposed to diffusion alone (Cserr et al., 1981; Groothuis et al., 2007), and arterial pulsations are also a driving force for perivascular transport (Rennels et al., 1985; Rennels et al., 1985). Intranasally applied drugs can move into perivascular spaces in the nasal passages or after reaching the brain and the widespread distribution observed within the CNS could be due to perivascular transport mechanisms (Thorne et al., 2004).
  • Pathways connecting the subarachnoid space containing CSF, perineurial spaces encompassing olfactory nerves, and the nasal lymphatics are important for CSF drainage and these same pathways provide access for intranasally applied therapeutics to the CSF and other areas of the CNS. Several studies document that tracers injected into the CSF in the cerebral ventricles or subarachnoid space drain to the underside of the olfactory bulbs into channels associated with olfactory nerves traversing the cribriform plate and reach the nasal lymphatic system and cervical lymph nodes (Bradbury et al., 1983; Hatterer et al., 2006; Johnston et al., 2004a); Kida et al., 1993; Walter et al., 2006a; Walter et al., 2006b). Drugs can access the CNS via these same pathways after intranasal administration, moving from the nasal passages to the CSF to the brain interstitial spaces and perivascular spaces for distribution throughout the brain. These drainage pathways are significant in a number of animal species (sheep, rabbits, and rats) accounting for approximately 50% of CSF clearance (Bradbury et al., 1981; Boulton et al., 1999; Boulton et al., 1996; Cserr et al., 1992). Pathways between the nasal passages and the CSF are still important and functional in humans, evidenced by the fact that therapeutics are directly delivered to the CSF following intranasal delivery, without entering the blood to an appreciable extent (Born et al., 2002). A number of intranasal studies demonstrate that drugs gain direct access to the CSF from the nasal cavity, followed by subsequent distribution to the brain and spinal cord. Many intranasally applied molecules rapidly enter the CSF, and this transport is dependent on the lipophilicity, molecular weight, and degree of ionization of the molecules (Dhanda et al., 2005; Born et al., 2002; Kumar et al., 1974; Sakane et al., 1995; Sakane et al., 1994; Wang et al., 2007). Assessing distribution into the CSF can provide information on the mechanism of intranasal delivery.
  • Optimal delivery to the CNS along neural pathways is associated with delivery of the agent to the upper third of the nasal cavity (Hanson et al., 2008). Although a supine position may be employed another position for targeting the olfactory region is with the “praying to Mecca” position, with the head down-and-forward. A supine position with the head angle at 70° or 90° may be suitable for efficient delivery to the CSF using a tube inserted into the nostrils to deliver the drug via intranasal administration (van den Berg et al., (2002)).
  • For intranasal drug administration nose drops may be administered over a period of 10-20 minutes to alternating nostrils every 1-2 minutes to allow the solution to be absorbed into the nasal epithelium (Thorne et al., 2004; Capsoni et al., 2002; Ross et al., 2004; Ross et al., 2008; Dhuria et al., 2009a; Dhuria et al., 2009b; Francis et al., 2008; Martinez et al., 2008). This noninvasive method does not involve inserting the device into the nostril. Instead, drops are placed at the opening of the nostril, allowing the individual to sniff the drop into the nasal cavity. Other administration methods in anesthetized individual involve sealing the esophagus and inserting a breathing tube into the trachea to prevent the nasal formulation from being swallowed and to eliminate issues related to respiratory distress (Chow et al., 1999; Chow et al., 2001; Fliedner et al., 2006; Dahlin et al., 2001). Flexible tubing can be inserted into the nostrils for localized delivery of a small volume of the drug solution to the respiratory or olfactory epithelia, depending on the length of the tubing (Chow et al., 1999; Van den Berg et al., 2003; van den Berg et al., 2004a; Banks et al., 2004; van den Berg et al., 2002; Vyas et al., 2006a; Charlton et al., 2007a; Gao et al., 2007a).
  • Nasal delivery devices, such as sprays, nose droppers or needle-less syringes, may be employed to target the agent to different regions of the nasal cavity. OptiMist™ is a breath actuated device that targets liquid or powder nasal formulations to the nasal cavity, including the olfactory region, without deposition in the lungs or esophagus (Djupesland et al., 2006). The ViaNase™ device can also be used to target a nasal spray to the olfactory and respiratory epithelia of the nasal cavity. Nasal drops tend to deposit on the nasal floor and are subjected to rapid mucociliary clearance, while nasal sprays are distributed to the middle meatus of the nasal mucosa (Scheibe et al., 2008).
  • The immune suppressant or immunotolerizing agent may be administered by any route including parenterally. In one embodiment, the immune suppressant or immunotolerizing agent may be administered by subcutaneous, intramuscular, or intravenous injection, orally, intrathecally, intracranially, or intranasally, or by sustained release, e.g., using a subcutaneous implant. The immune suppressant or immunotolerizing agent may be dissolved or dispersed in a liquid carrier vehicle. For parenteral administration, the active material may be suitably admixed with an acceptable vehicle, e.g., of the vegetable oil variety such as peanut oil, cottonseed oil and the like. Other parenteral vehicles such as organic compositions using solketal, glycerol, formal, and aqueous parenteral formulations may also be used. For parenteral application by injection, compositions may comprise an aqueous solution of a water soluble pharmaceutically acceptable salt of the active acids according to the invention, desirably in a concentration of 0.01-10%, and optionally also a stabilizing agent and/or buffer substances in aqueous solution. Dosage units of the solution may advantageously be enclosed in ampules.
  • The composition, e.g., rAAV containing composition, immune suppressant containing composition or immunotolerizing composition, may be in the form of an injectable unit dose. Examples of carriers or diluents usable for preparing such injectable doses include diluents such as water, ethyl alcohol, macrogol, propylene glycol, ethoxylated isostearyl alcohol, polyoxyisostearyl alcohol and polyoxyethylene sorbitan fatty acid esters, pH adjusting agents or buffers such as sodium citrate, sodium acetate and sodium phosphate, stabilizers such as sodium pyrosulfite, EDTA, thioglycolic acid and thiolactic acid, isotonic agents such as sodium chloride and glucose, local anesthetics such as procaine hydrochloride and lidocaine hydrochloride. Furthermore usual solubilizing agents and analgesics may be added. Injections can be prepared by adding such carriers to the enzyme or other active, following procedures well known to those of skill in the art. A thorough discussion of pharmaceutically acceptable excipients is available in REMINGTON'S PHARMACEUTICAL SCIENCES (Mack Pub. Co., N.J. 1991). The pharmaceutically acceptable formulations can easily be suspended in aqueous vehicles and introduced through conventional hypodermic needles or using infusion pumps. Prior to introduction, the formulations can be sterilized with, preferably, gamma radiation or electron beam sterilization.
  • When the immune suppressant or immunotolerizing agent is administered in the form of a subcutaneous implant, the compound is suspended or dissolved in a slowly dispersed material known to those skilled in the art, or administered in a device which slowly releases the active material through the use of a constant driving force such as an osmotic pump. In such cases, administration over an extended period of time is possible.
  • The dosage at which the immune suppressant or immunotolerizing agent containing composition is administered may vary within a wide range and will depend on various factors such as the severity of the disease, the age of the patient, etc., and may have to be individually adjusted. A possible range for the amount which may be administered per day is about 0.1 mg to about 2000 mg or about 1 mg to about 2000 mg. The compositions containing the immune suppressant or immunotolerizing agent may suitably be formulated so that they provide doses within these ranges, either as single dosage units or as multiple dosage units. In addition to containing an immune suppressant, the subject formulations may contain one or more rAAV encoding a therapeutic gene product.
  • Compositions described herein may be employed in combination with another medicament. The compositions can appear in conventional forms, for example, aerosols, solutions, suspensions, or topical applications, or in lyophilized form.
  • Typical compositions include a rAAV, an immune suppressant, a permeation enhancer, or a combination thereof, and a pharmaceutically acceptable excipient which can be a carrier or a diluent. For example, the active agent(s) may be mixed with a carrier, or diluted by a carrier, or enclosed within a carrier. When the active agent is mixed with a carrier, or when the carrier serves as a diluent, it can be solid, semi-solid, or liquid material that acts as a vehicle, excipient, or medium for the active agent. Some examples of suitable carriers are water, salt solutions, alcohols, polyethylene glycols, polyhydroxyethoxylated castor oil, peanut oil, olive oil, gelatin, lactose, terra alba, sucrose, dextrin, magnesium carbonate, sugar, cyclodextrin, amylose, magnesium stearate, talc, gelatin, agar, pectin, acacia, stearic acid or lower alkyl ethers of cellulose, silicic acid, fatty acids, fatty acid amines, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, polyoxyethylene, hydroxymethylcellulose and polyvinylpyrrolidone. Similarly, the carrier or diluent can include any sustained release material known in the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax.
  • The formulations can be mixed with auxiliary agents which do not deleteriously react with the active agent(s). Such additives can include wetting agents, emulsifying and suspending agents, salt for influencing osmotic pressure, buffers and/or coloring substances preserving agents, sweetening agents or flavoring agents. The compositions can also be sterilized if desired.
  • If a liquid carrier is used, the preparation can be in the form of a liquid such as an aqueous liquid suspension or solution. Acceptable solvents or vehicles include sterilized water, Ringer's solution, or an isotonic aqueous saline solution.
  • The agent(s) may be provided as a powder suitable for reconstitution with an appropriate solution as described above. Examples of these include, but are not limited to, freeze dried, rotary dried or spray dried powders, amorphous powders, granules, precipitates, or particulates. The composition can optionally contain stabilizers, pH modifiers, surfactants, bioavailability modifiers and combinations of these. A unit dosage form can be in individual containers or in multi-dose containers.
  • Compositions contemplated by the present invention may include, for example, micelles or liposomes, or some other encapsulated form, or can be administered in an extended release form to provide a prolonged storage and/or delivery effect, e.g., using biodegradable polymers, e.g., polylactide-polyglycolide. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Polymeric nanoparticles, e.g., comprised of a hydrophobic core of polylactic acid (PLA) and a hydrophilic shell of methoxy-poly(ethylene glycol) (MPEG), may have improved solubility and targeting to the CNS. Regional differences in targeting between the microemulsion and nanoparticle formulations may be due to differences in particle size.
  • Liposomes are very simple structures consisting of one or more lipid bilayers of amphiphilic lipids, i.e., phospholipids or cholesterol. The lipophilic moiety of the bilayers is turned towards each other and creates an inner hydrophobic environment in the membrane. Liposomes are suitable drug carriers for some lipophilic drugs which can be associated with the non-polar parts of lipid bilayers if they fit in size and geometry. The size of liposomes varies from 20 nm to few μm.
  • Mixed micelles are efficient detergent structures which are composed of bile salts, phospholipids, tri, di- and monoglycerides, fatty acids, free cholesterol and fat soluble micronutrients. As long-chain phospholipids are known to form bilayers when dispersed in water, the preferred phase of short chain analogues is the spherical micellar phase. A micellar solution is a thermodynamically stable system formed spontaneously in water and organic solvents. The interaction between micelles and hydrophobic/lipophilic drugs leads to the formation of mixed micelles (MM), often called swallen micelles, too. In the human body, they incorporate hydrophobic compounds with low aqueous solubility and act as a reservoir for products of digestion, e.g. monoglycerides.
  • Lipid microparticles includes lipid nano- and microspheres. Microspheres are generally defined as small spherical particles made of any material which are sized from about 0.2 to 100 μm. Smaller spheres below 200 nm are usually called nanospheres. Lipid microspheres are homogeneous oil/water microemulsions similar to commercially available fat emulsions, and are prepared by an intensive sonication procedure or high pressure emulsifying methods (grinding methods). The natural surfactant lecithin lowers the surface tension of the liquid, thus acting as an emulsifier to form a stable emulsion. The structure and composition of lipid nanospheres is similar to those of lipid microspheres, but with a smaller diameter.
  • Polymeric nanoparticles serve as carriers for a broad variety of ingredients. The active components may be either dissolved in the polymetric matrix or entrapped or adsorbed onto the particle surface. Polymers suitable for the preparation of organic nanoparticles include cellulose derivatives and polyesters such as poly(lactic acid), poly(glycolic acid) and their copolymer. Due to their small size, their large surface area/volume ratio and the possibility of functionalization of the interface, polymeric nanoparticles are ideal carrier and release systems. If the particle size is below 50 nm, they are no longer recognized as particles by many biological and also synthetic barrier layers, but act similar to molecularly disperse systems.
  • Thus, the composition of the invention can be formulated to provide quick, sustained, controlled, or delayed release, or any combination thereof, of the active agent after administration to the individual by employing procedures well known in the art. In one embodiment, the enzyme is in an isotonic or hypotonic solution. In one embodiment, for enzymes that are not water soluble, a lipid based delivery vehicle may be employed, e.g., a microemulsion such as that described in WO 2008/049588, the disclosure of which is incorporated by reference herein, or liposomes.
  • In one embodiment, the preparation can contain an agent, dissolved or suspended in a liquid carrier, such as an aqueous carrier, for aerosol application. The carrier can contain additives such as solubilizing agents, e.g., propylene glycol, surfactants, absorption enhancers such as lecithin (phosphatidylcholine) or cyclodextrin, or preservatives such as parabens. For example, in addition to solubility, efficient delivery to the CNS following intranasal administration may be dependent on membrane permeability. For enzymes where paracellular transport is hindered due to size and polarity, improving membrane permeability may enhance extracellular mechanisms of transport to the CNS along olfactory and trigeminal nerves. One approach to modifying membrane permeability within the nasal epithelium is by using permeation enhancers, such as surfactants, e.g., lauroylcarnitine (LC), bile salts, lipids, cyclodextrins, polymers, or tight junction modifiers.
  • Generally, the active agents are dispensed in unit dosage form including the active ingredient together with a pharmaceutically acceptable carrier per unit dosage. Usually, dosage forms suitable for nasal administration include from about 125 lug to about 125 mg, e.g., from about 250 lug to about 50 mg, or from about 2.5 mg to about 25 mg, of the compounds admixed with a pharmaceutically acceptable carrier or diluent.
  • Dosage forms can be administered daily, or more than once a day, such as twice or thrice daily. Alternatively dosage forms can be administered less frequently than daily, such as every other day, or weekly, if found to be advisable by a prescribing physician.
  • The invention will be described by the following non-limiting examples.
  • Example I AAV Vector-Mediated Iduronidase Gene Delivery in a Murine Model of Mucopolysaccharidosis Type I: Comparing Different Routes of Delivery to the CNS
  • Mucopolysaccharidosis type I (MPS I) is an inherited metabolic disorder caused by deficiency of the lysosomal enzyme alpha-L-iduronidase (IDUA). Systemic and abnormal accumulation of glycosaminoglycans is associated with growth delay, organomegaly, skeletal dysplasia, and cardiopulmonary disease. Individuals with the most severe form of the disease (Hurler syndrome) suffer from neurodegeneration, mental retardation, and early death. The two current treatments for MPS I (hematopoietic stem cell transplantation and enzyme replacement therapy) cannot effectively treat all central nervous system (CNS) manifestations of the disease.
  • With respect to gene therapy, it was previously demonstrated that intravascular delivery of AAV-9 in adult mice does not achieve widespread direct neuronal targeting (see Foust et al, 2009). Previous work also showed that direct injection of AAV8-IDUA into the CNS of adult IDUA-deficient mice resulted in a low frequency or a poor level of transgene expression (see FIG. 18). The following examples, which use a pre-clinical model for the treatment of MPS1, surprisingly demonstrate that direct injection of AAV9-IDUA into the CNS of immunocompetent adult IDUA-deficient mice resulted in IDUA enzyme expression and activity that is the same or higher than IDUA enzyme expression and activity in wild-type adult mice (see FIG. 15, infra).
  • Methods
  • AAV9-IDUA preparation. The AAV-IDUA vector construct (MCI) has been previously described (Wolf et al., 2011) (mCags promoter). AAV-IDUA plasmid DNA was packaged into AAV9 virions at the University of Florida Vector Core, yielding a titer of 3×1013 vector genomes per milliliter.
  • ICV infusions. Adult Idua−/− mice were anesthetized using a cocktail of ketamine and xylazine (100 mg ketamine+10 mg xylazine per kg) and placed on a stereotactic frame. Ten microliters of AAV9-IDUA were infused into the right-side lateral ventricle (stereotactic coordinates AP 0.4, ML 0.8, DV 2.4 mm from bregma) using a Hamilton syringe. The animals were returned to their cages on heating pads for recovery.
  • Intrathecal infusions. Infusions into young adult mice were carried out by injection of 10 μL AAV vector containing solution between the L5 and L6 vertebrae 20 minutes after intravenous injection of 0.2 mL 25% mannitol.
  • Immunotolerization. Newborn IDUA deficient mice were injected through the facial temporal vein with 5 μL containing 5.8 μg of recombinant iduronidase protein (Aldurazyme), and then the animals were returned to their cage.
  • Cyclophosphamide immunosuppression. For immunosuppression, animals were administered cyclophosphamide once per week at a dose of 120 mg/kg starting one day after infusion with AAV9-IDUA vector.
  • Animals. Animals were anesthetized with ketamine/xylazine (100 mg ketamine+10 mg xylazine per kg) and transcardially perfused with 70 mL PBS prior to sacrifice. Brains were harvested and microdissected on ice into cerebellum, hippocampus, striatum, cortex, and brainstem/thalamus (“rest”). The samples were frozen on dry ice and then stored at −80° C. Samples were thawed and homogenized in 1 mL of PBS using a motorized pestle and permeabilized with 0.1% Triton X-100. IDUA activity was determined by fluorometric assay using 4MU-iduronide as the substrate. Activity is expressed in units (percent substrate converted to product per minute) per mg protein as determined by Bradford assay (Bio Rad).
  • Tissues. Tissue homogenates were clarified by centrifugation for 3 minutes at 13,000 rpm using an Eppendorf tabletop centrifuge model 5415D (Eppendorf) and incubated overnight with proteinase K, DNase1, and Rnase. GAG concentration was determined using the Blyscan Sulfated Glycosaminoglycan Assay (Accurate Chemical) according to the manufacturer's instructions.
  • Results
  • FIG. 1 shows the results for iduronidase-deficient mice that were administered MV either intracerebroventricularly (ICV) or intrathecally (IT). To prevent immune response, animals were either immunosuppressed with cyclophosphamide (CP), immunotolerized at birth by intravenous administration of human iduonidase protein (aldurazyme), or the injections were carried out in NOD-SCID immunodeficient mice that were also iduronidase deficient. Animals were sacrificed at the indicated time post-treatment, the brains were microdissected and extracts assayed for iduronidase activity.
  • FIG. 2 illustrates data for immunodeficient, IDUA deficient animals injected ICV with AAV-IDUA vector. Those animals exhibited high levels of IDUA expression (10 to 100 times wild type) in all areas of the brain, with the highest level observed in the brain stem and thalamus (“rest”).
  • Immunosuppressed animals administered AAV vector by ICV route had a relatively lower level of enzyme in the brain compared to the immunodeficient animals (FIG. 3). Note that immunosuppression may have been compromised in these animals because CP was withdrawn 2 weeks before sacrifice due to poor health.
  • FIG. 4 shows data for immunosuppressed animals administered AAV vector by the IT route. Immunotolerized animals administered AAV vector ICV exhibited widespread IDUA activity in all parts of the brain (FIG. 5), similar to that observed in the immunodeficient animals, indicating the effectiveness of the immunotolerization procedure.
  • FIG. 6 is a compilation of all mean levels of IDUA activity for side-by-side comparison, and FIG. 7 is data grouped according the area of the brain.
  • GAG storage material was assayed in the different sections of the brain for all four of the test groups. For each group, the mean of each portion of the brain is shown on the left, the values for each of the individual animals is shown on the right (FIG. 8). IDUA deficient animals (far left) contained high levels of GAG compared to wild type animals (magenta bar). GAG levels were at wild-type or lower than wild type for all portions of the brain in all groups of AAV-treated animals. GAG levels were slightly although not significantly higher than wild-type in cortex and brainstem of animals administered AAV9-IDUA intrathecally.
  • Conclusions
  • The results show high and widespread distribution of IDUA in the brain regardless of the route of delivery (ICV or IT) although IDUA expression in striatum and hippocampus was lower in animals injected IT versus ICV. There appears to be an immune response since immune deficient mice have higher levels of expression than immunocompetent mice. With regard to ICV injection, when CP was withdrawn early, IDUA expression is lower. In addition, immunotolerization was effective in restoring high levels of enzyme activity. Further, GAG levels were restored to normal in all treated experimental groups of mice.
  • Example II Methods
  • AAV9IDUA Preparation. AAV-IDUA plasmid was packaged into AAV9 virions at either the University of Florida vector core, or the University of Pennsylvania vector core, yielding a titer of 1-3×1013 vector genomes per millilter.
  • ICV infusions. See Example I.
  • Intrathecal infusions. See Example I.
  • Immunotolerization. As in Example I except: for multiple tolerizations, newborn IDUA deficient mice were injected with the first dose of Aldurazyme in the facial temporal vein, followed by 6 weekly injections administered intraperitoneally.
  • Cyclophosphamide immunosuppression. See Example I.
  • Animals. Animals were anesthetized with ketamine/xylazine (100 mg ketamine+10 mg xylazine per kg) and transcardially perfused with 70 mL PBS prior to sacrifice. Brains were harvested and microdissected on ice into cerebellum, hippocampus, striatum, cortex, and brainstem/thalamus (“rest”). The samples were frozen on dry ice and then stored at −80° C.
  • Tissue IDUA activity. Tissue samples were thawed and homogenized in saline in a tissue homogenizer. Tissue homogenates were clarified by centrifugation at 15,000 rpm in a benchtop Eppendorf centrifuge at 4° C. for 15 minutes. Tissue lysates (supernatant) were collected and analyzed for IDUA activity and GAG storage levels.
  • Tissue GAG levels. Tissue lysates were incubated overnight with Proteinase K, RNase and DNase. GAG levels were analyzed using the Blyscan Sulfated Glycosaminoglycan Assay according to the manufacturer's instructions.
  • IDUA Vector copies. Tissue homogenates were used for DNA isolation and subsequent QPCR, as described in Wolf et al. (2011).
  • Results
  • FIG. 9 illustrates the experimental design and groups. Animals were administered AAV9IDUA vector either by intracerebroventricular (ICV) or intrathecal (IT) infusion. Vector administration was carried out in NOD-SCID immunodeficient (ID) mice that were also IDUA deficient, or in IDUA deficient mice that were either immunosuppressed with cyclophosphamide (CP), or immunotolerized at birth by a single or multiple injections of human iduronidase protein (Aldurazyme). The times of treatment with vector and sacrifice are as indicated in FIG. 9. All vector administrations were carried out in adult animals ranging in age from 3-4.5 months. Animals were injected with 10 μL of vector at a dose of 3×1011 vector genomes per 10 microliters.
  • FIG. 10 shows IDUA enzyme activities in intracranially infused, immunodeficient, IDUA deficient mice. High levels of enzyme activity were seen in all areas of the brain, ranging from 30- to 300-fold higher than wild type levels. Highest enzyme expressions were seen in thalamus and brain stem, and in the hippocampus.
  • Animals that were injected intracranially and immunosuppressed with cyclophosphamide (CP) demonstrated significantly lower levels of enzyme activity than other groups (FIG. 11). However, CP administration in this case had to be withdrawn 2 weeks prior to sacrifice due to poor health of the animals.
  • IDUA enzyme levels in animals tolerized at birth with IDUA protein (Aldurazyme) and administered vector intracranially are depicted in FIG. 12. All animals showed high enzyme levels in all parts of the brain that ranged from 10- to 1000-fold higher than wild type levels, similar to levels achieved in immunodeficient animals, indicating the effectiveness of the immunotolerization procedure.
  • FIG. 13 depicts IDUA enzyme levels in mice that were injected intrathecally and administered CP on a weekly basis. Elevated levels of IDUA were observed in all parts of the brain, especially in the cerebellum and the spinal cord. Levels of enzyme were the lowest in the striatum and hippocampus with activities at wild type levels.
  • IDUA deficient mice were tolerized with Aldurazyme as described, and injected with vector intrathecally (FIG. 14). There was widespread IDUA enzyme activity in all parts of the brain, with highest levels of activity in the brain stem and thalamus, olfactory bulb, spinal cord and the cerebellum. Similar to the data in FIG. 13, the lowest levels of enzyme activity were seen in the striatum, cortex and hippocampus.
  • Control immunocompetent IDUA deficient animals were infused with vector intrathecally, without immunosuppression or immunotolerization (FIG. 15). The results indicate that although enzyme activities were at wild type levels or slightly higher, they are significantly lower than what was observed in animals that underwent immunomodulation. The decreases in enzyme levels were especially significant in the cerebellum, olfactory bulb and thalamus and brain stem, areas that expressed the highest levels of enzyme in immunomodulated animals.
  • Animals were assayed for GAG storage material, as shown in FIG. 15. All groups demonstrated clearance of GAG storage, with GAG levels similar to that observed in wild type animals. Animals that were immunosuppressed and injected with AAV-IDUA vector intrathecally had GAG levels in the cortex that were slightly higher than wild type, but still much lower than untreated IDUA deficient mice.
  • The presence of AAV9IDUA vector in animals that were immunotolerized and injected with vector either intracranially or intrathecally was evaluated by QPCR, as illustrated in FIG. 16. IDUA copies per cell were higher in animals infused intracranially in comparison with animals infused intrathecally, which is consistent with the higher level of enzyme activity seen in animals injected intracranially.
  • Conclusions
  • High, widespread, and therapeutic levels of IDUA were observed in all areas of the brain after intracerebroventricular and intrathecal routes of AAV9IDUA administration in adult mice Enzyme activities were restored to wild type levels or slightly higher in immunocompetent IDUA deficient animals infused with AAV-IDUA intrathecally. Significantly higher levels of IDUA enzyme were observed for both routes of vector injection in animals immunotolerized starting at birth by administration of IDUA protein.
  • Example III
  • Adult immunocompetent IDUA deficient mice (12 weeks old) were anesthetized with ketamine/xylazine, followed by intranasal infusion of AAV9IDUA vector. Vector was administered by applying eight 3 μL drops with a micropipette to the intranasal cavity, alternating between nostrils, at 2 minute intervals between each application. A total of 2.4-7×1011 vector genomes was administered to each adult animal, depending on source of vector. Animals were immunosuppressed with 120 mg/kg cyclophosphamide administered weekly, starting the day after vector administration. Mice were sacrificed at 12 weeks post vector infusion, animals were assayed for IDUA enzyme expression and vector copies in the brain.
  • REFERENCES
    • Al-Ghananeem et al., AAPS Pharm. Sci. Tech., 3:E5 (2002).
    • Bagger et al., Eur. J. Pharm. Sci., 21:235-242 (2004b).
    • Bagger et al., Int. J. Pharm., 269:311-322 (2004a).
    • Baker et al., Exp. Brain Res., 63:461 (1986).
    • Balin et al., J. Comp. Neurol., 251:260-280 (1986).
    • Banks et al., J. Drug Target, 17:91-97 (2009).
    • Banks et al., J. Pharmacol. Exp. Ther., 309:469 (2004).
    • Banks, Biopolymers, 90:589 (2008).
    • Barakat et al., J. Pharm. Pharmacol., 58:63 (2006).
    • Barbier et al., Mol. Genet. Metab., 110:303 (2013).
    • Baumgartner et al., Neuron., 58:639 (2008).
    • Benedict et al., Neuroendocrinology, 86:136 (2007b).
    • Benedict et al., Neuropsychopharmacology, 32:239 (2007a).
    • Benedict et al., Psychoneuroendocrinology, 29:1326 (2004).
    • Bjoraker et al., J. Dev. Behay. Ped., 27:290 (2006).
    • Blits et al., J. Neuros. Methods, 185:257 (2010).
    • Born et al., Nat. Neurosci., 5:514 (2002).
    • Boulton et al., Am. J. Physiol., 276:R818 (1999).
    • Boulton et al., Neuropathol. Appl. Neurobiol., 22:325 (1996).
    • Bradbury et al., Am. J. Physiol., 240:F329 (1981).
    • Bradbury et al., J. Physiol., 339:519 (1983).
    • Brady, Ann. Rev. Med., 57:283 (2006).
    • Broadwell et al., J. Comp. Neurol., 242:632 (1985).
    • Broekman et al., Neuroscience, 138:501 (2006).
    • Buck, In: Kandel E R, Schwartz J H, Jessell T M, editors. Principles of neural science. 4th edition. New York: McGraw-Hill Companies. pp. 625-652 (2000).
    • Buxer et al., J. Neurochem., 56:1012 (1991).
    • Cai et al., Sichuan Da Xue Xue Bao Yi Xue Ban, 39:438 (2008).
    • Capsoni et al., Proc. Natl. Acad. Sci. USA, 99:12432 (2002).
    • Carare et al., Neuropathol. Appl. Neurobiol., 34:131 (2008).
    • Cauna, In: Proctor D F, Andersen I, editors. Amsterdam: Elsevier Biomedical Press. pp. 45-69 (1982).
    • Charlton et al., Int. J. Pharm., 338:94 (2007b).
    • Charlton et al., J. Drug Target, 15:370 (2007a).
    • Charlton et al., Pharm. Res., 25:1531 (2008).
    • Chen et al., J. Alzheimers Dis., 1:35 (1998).
    • Chen et al., J. Pharm. Sci., 95:1364 (2006).
    • Chow et al., J. Pharm. Sci., 88:754 (1999).
    • Chow et al., J. Pharm. Sci., 90:1729 (2001).
    • Clerico et al., In: Doty R L, editor. Handbook of olfaction and gustation. 2nd edition. New York: Marcel Dekker, Inc. pp. 1-16 (2003).
    • Costantino et al., Int. J. Pharm., 337:1 (2007).
    • Cserr et al., Am. J. Physiol., 240:F319 (1981).
    • Cserr et al., Brain Pathol., 2:269 (1992).
    • Dahlin et al., Eur. J. Pharm. Sci., 14:75 (2001).
    • Danhof et al., American Association of Pharmaceutical Scientists Annual Meeting, Atlanta, Ga. (2008).
    • Danielyan et al., Eur. J. Cell. Biol., 88:315 (2009).
    • Davis et al., Clin. Pharmacokinet., 42:1107 (2003).
    • de Lorenzo, In: Wolstenholme GEW, Knight J, editors. Taste and smell in vertebrates. London: Churchill. pp. 151-175 (1970).
    • De Rosa et al., Proc. Natl. Acad. Sci. USA, 102:3811 (2005).
    • DeSesso, Qual. Assur., 2:213 (1993).
    • deSouza et al., Eur. Neuropsychopharmacol., 19:53 (2009).
    • Dhanda et al., Drug Del. Tech., 5:64 (2005).
    • Dhuria et al., J. Pharm. Sci., 98:2501 (2009b).
    • Dhuria et al., J. Pharmaceutical Sciences, 99:1654 (2010).
    • Dhuria et al., J. Pharmacol. Exp. Ther., 328:312 (2009a).
    • Diano et al., J. Clin. Invest., 118:26 (2008).
    • Dickson et al., Mol. Gen. Metab., 91:61 (2007).
    • Djupesland et al., Laryngoscope, 116:466 (2006).
    • Domes et al., Biol. Psychiatry, 61:731 (2007b).
    • Domes et al., Biol. Psychiatry, 62:1187 (2007a).
    • Dufes et al., Int. J. Pharm., 255:87 (2003).
    • Einer-Jensen et al., Exp. Brain Res., 130:216 (2000b).
    • Einer-Jensen et al., Pharmacol. Toxicol., 87:276 (2000a).
    • Einer-Jensen et al., Reproduction, 129:9 (2005).
    • Ellinwood et al., Mol. Genet. Metab., 91:239 (2007).
    • Fehm et al., J. Clin. Endocrinol. Metab., 86:1144 (2001).
    • Field et al., J. Neurocytol., 32:317 (2003).
    • Fliedner et al., Endocrinology., 17:2088 (2006).
    • Foust et al., Nat. Biotech., 27:5 (2009)).
    • Francis et al., Brain, 131:3311 (2008).
    • Fratantoni et al., Science, 162:570 (1968).
    • Frey et al., Drug Delivery, 4:87 (1997).
    • Frey I I, Drug Del. Tech., 2:46 (2002).
    • Fuss et al., Eur. J. Neurosci., 22:2649 (2005).
    • Gao et al., Biomaterials, 27:3482 (2006).
    • Gao et al., Int. J. Pharm., 340:207 (2007a).
    • Gao et al., J. Control Release, 121:156 (2007b).
    • Gopinath et al., Current Ther. Res., 23:596 (1978).
    • Gozes et al., Curr. Alzheimer Res., 4:507 (2007).
    • Graff et al., Pharm. Res., 20:1225 (2003).
    • Graff et al., Pharm. Res., 22:235 (2005a).
    • Graff et al., Pharm. Res., 22:86 (2005b).
    • Gray, 15th revised edition (Classic Collectors edition). New York: Bounty Books (1978).
    • Grevers et al., Arch. Otorhinolaryngol., 244:55 (1987).
    • Groothuis et al., J. Cereb. Blood Flow Metab., 27:43 (2007).
    • Gross et al., J. Anat., 135:83 (1982).
    • Guastella et al., Biol. Psychiatry, 63:3 (2008).
    • Hadaczek et al., Mol. Ther., 14:69 (2006).
    • Hallschmid et al., Regul. Pept., 149:79 (2008).
    • Han et al., J. Mol. Med., 85:75 (2007).
    • Hanson et al., BMC Neurosci., 9:S5 (2008).
    • Hanson et al., Drug Del. Tech., 4:66 (2004).
    • Hanson et al., San Diego, Calif.: Society for Neuroscience (2007).
    • Hanson et al., In: EPO, editor. Biopharm and HealthPartners Research Foundation (2008).
    • Hartung et al., J. Am. Soc. Gene Ther., 9:866 (2004).
    • Hartung et al., Mol. Thera., 9:869 (2004).
    • Hashizume et al., Neuro. Oncol., 10:112 (2008).
    • Hatterer et al., Blood, 107:806 (2006).
    • Herati et al., J. Gene Med., 10:972 (2008).
    • Hess et al., Exp. Neuro., 186:134 (2004).
    • Horvat et al., Eur. J. Pharm. Biopharm., 72:252 (2009).
    • Hussar et al., Chem. Senses, 27:7 (2002).
    • Ilium, J. Control Release, 87:187 (2003).
    • Ilium, J. Pharm. Pharmacol., 56:3 (2004).
    • Itaya et al., Brain Res., 398:397 (1986).
    • Jansson et al., J. Drug Target, 10:379 (2002).
    • Jogani et al., Alzheimer Dis. Assoc. Disord., 22:116 (2008).
    • Johnston et al., Cerebrospinal Fluid Res., 1:2 (2004).
    • Kakkis et al., Mol. Gen. Met., 83:163 (2004).
    • Kandimalla et al., J. Pharm. Sci., 94:613 (2005b).
    • Kandimalla et al., Pharm. Res., 22:1121 (2005a).
    • Kida et al., Neuropathol. Appl. Neurobiol., 19:480 (1993).
    • Kirsch et al., J. Neurosci., 25:11489 (2005).
    • Klein et al., J. Am. Soc. Gene Ther., 13:517 (2006).
    • Koos et al., Neuroreport, 16:1929 (2005).
    • Kosfeld et al., Nature, 435:673 (2005).
    • Kristensson et al., Acta Neuropathol (Berl), 19:145 (1971).
    • Krivit, Springer Seminars in Immunopathology, 26:119 (2004).
    • Kumar et al., Curr. Sci., 43:435 (1974).
    • Kumar et al., Int. J. Pharm., 358:285 (2008).
    • Li et al., Chin. J. Physiol., 48:7 (2005c).
    • Li et al., Glia, 52:245 (2005a).
    • Li et al., J. Neurocytol., 34:343 (2005b).
    • Loftus et al., Neuroscience, 139:1061 (2006).
    • Luzzati et al., J. Mol. Biol., 343:199 (2004).
    • Mackay-Sim, In: Doty RL, editor. Handbook of olfaction and gustation. 2nd edition. New York: Marcel Dekker, Inc. pp. 93-113 (2003).
    • Martinez et al., Neuroscience, 157:908 (2008).
    • Minn et al., J. Drug Target, 10:285 (2002).
    • Miragall et al., J. Comp. Neurol., 341:433 (1994).
    • Muenzer, J. Pediatrics, 144:S27 (2004).
    • Munoz-Rojas et al., Am. J. Med. Gen., 146A:2538 (2008).
    • Neufeld and Muenzer, In A. L. B. C. R. Scriver, W. S. Sly, et al (ed.), McGraw Hill, NY, pg. 3421 (2001).
    • Nonaka et al., J. Pharmacol. Exp. Ther., 325:513 (2008).
    • Ohlfest et al., Blood, 105:2691 (2005).
    • Orchard et al., J. Pediatrics, 151:340 (2007).
    • Owens et al., Diabet. Med., 20:886 (2003).
    • Pan et al., Brain Res., 1188:241 (2008).
    • Pardridge, NeuroRx, 2:3 (2005).
    • Parker et al., Psychoneuroendocrinology, 30:924 (2005).
    • Pastores, Exp. Opin. Biol. Ther., 8:1003 (2008).
    • Perl et al., Lancet, 1:1028 (1987).
    • Peters et al., Bone Marrow Transpl., 31:229 (2003).
    • Pollock et al., J. Anat., 191:337 (1997).
    • Raghavan et al., J. Laryngol. Otol., 114:456 (2000).
    • Reger et al., J. Alzheimers Dis., 13:323 (2008a).
    • Reger et al., Neurobiol. Aging, 27:451 (2006).
    • Reger et al., Neurology, 70:440 (2008b).
    • Rennels et al., Adv. Neurol., 52:431 (1990).
    • Rennels et al., Brain Res., 326:47 (1985).
    • Reolon et al., Brain Res., 1076:225 (2006).
    • Rimmele et al., J. Neurosci., 29:38 (2009).
    • Ross et al., J. Neuroimmunol., 151:66 (2004).
    • Ross et al., Neurosci. Lett., 439: 30 (2008).
    • Sakane et al., J. Pharm. Pharmacol., 46:378 (1994).
    • Sakane et al., J. Pharm. Pharmacol., 47:379 (1995).
    • Sarkar, Pharm. Res., 9:1 (1992).
    • Schaefer et al., J. Comp. Neurol., 444:221 (2002).
    • Scheibe et al., Arch. Otolaryngol. Head Neck Surg., 134:643 (2008).
    • Schley et al., J. Theor. Biol., 238:962 (2006).
    • Schulz et al., Endocrinology, 145:2696 (2004).
    • Scott et al., Am. J. Hum. Genet., 53:973 (1993).
    • Shimizu et al., Int. J. Obes. (Lond), 29:858 (2005).
    • Shipley, Brain Res. Bull., 15:129 (1985).
    • Skipor et al., Reprod. Biol., 3:143 (2003).
    • Steen et al., J. Alzheimers Dis., 7:63 (2005).
    • Stefanczyk-Krzymowska et al., Exp. Physiol., 85:801 (2000).
    • Takano et al., J. Histochem. Cytochem., 53:611 (2005).
    • Thorne et al., Brain Res., 692:278 (1995).
    • Thorne et al., Clin. Pharmacokinet., 40:907 (2001).
    • Thorne et al., Neuroscience, 127:481 (2004).
    • Thorne et al., Neuroscience, 152:785 (2008).
    • Thorne, R G. 2002. The nasal pathways for drug delivery to the central nervous system: Studies with protein tracers and therapeutics. Doctoral Dissertation, University of Minnesota.
    • Unger et al., J. Neuropath. Exp. Neuro., 52:460 (1993).
    • van den Berg et al., Eur. J. Pharm. Biopharm., 58:131 (2004b).
    • Van den Berg et al., J. Drug Target, 11:325 (2003).
    • van den Berg et al., J. Neurosci. Methods, 116:99 (2002).
    • van den Berg et al., Pharm. Res., 21:799 (2004a).
    • Van Diest et al., J. Anat., 128:293 (1979).
    • Vyas et al., AAPS PharmSciTech., 7:E1 (2006c).
    • Vyas et al., Crit. Rev. Ther. Drug Carrier Syst., 23:319 (2006b).
    • Vyas et al., J. Drug Target, 13:317 (2005).
    • Vyas et al., J. Pharm. Sci., 95:570 (2006a).
    • Walter et al., Arch. Histol. Cytol., 69:37 (2006a).
    • Walter et al., Neuropathol. Appl. Neurobiol., 32:388 (2006b).
    • Wang et al., Cancer Chemother. Pharmacol., 57:97 (2006a).
    • Wang et al., Eur. J. Pharm. Biopharm., 70:735 (2008).
    • Wang et al., Int. J. Pharm., 317:40 (2006b).
    • Wang et al., Int. J. Pharm., 341:20 (2007).
    • Watson et al., Gene Therapy, 16 Feb. 2006, doi:10.1038/sj.gt.3302735.
    • Weller et al., Neurol. Res., 25:611 (2003).
    • Westin et al., Eur. J. Pharm. Sci., 24:565 (2005).
    • Westin et al., Pharm. Res., 23:565 (2006).
    • Williams et al., J. Comp. Neurol., 470:50 (2004).
    • Wioland et al., J. Histochem. Cytochem., 48:1215 (2000).
    • Wolf et al., Neurobio. Dis., 43:123 (2011).
    • Xu et al., J. Clin. Invest., 118:272 (2008).
    • Yamada et al., Am. J. Physiol., 261:H1197 (1991).
    • Yang et al., J. Pharm. Sci., 94:1577 (2005).
    • Zhang et al., Acta Neuropathol. (Berl), 83:233 (1992).
    • Zhang et al., Acta Pharmacol. Sin., 25:522 (2004a).
    • Zhang et al., Int. J. Pharm., 275:85 (2004b).
    • Zhang et al., J. Drug Target, 14:281 (2006).
    • Zhao et al., Acta Pharmacol. Sin., 28:273 (2007).
    • Zhao et al., Chin. Med. Sci. J., 19:257 (2004).
    • Zheng et al., Mol. Genet. Metab., 79:233 (2003).
    • Ziegler and Shapiro, In J. Donders and S. Hunter (ed.), Cambridge University Press, p. 427 (2007).
  • All publications, patents and patent applications are incorporated herein by reference. While in the foregoing specification, this invention has been described in relation to certain preferred embodiments thereof, and many details have been set forth for purposes of illustration, it will be apparent to those skilled in the art that the invention is susceptible to additional embodiments and that certain of the details herein may be varied considerably without departing from the basic principles of the invention.

Claims (26)

What is claimed is:
1. A method to inhibit or treat one or more symptoms of Pompe Disease in a human in need thereof, consisting of:
administering to the human an amount of a recombinant adeno-associated virus (rAAV) vector having an AAV8, AAV9 or AAVrh10 capsid encapsidating a rAAV genome having an open reading frame encoding acid maltase that is effective to inhibit or treat the one or more symptoms of Pompe Disease in the human.
2. The method of claim 1 wherein the human is an immunocompetent adult.
3. The method of claim 1 wherein neurodegeneration is inhibited or treated by the administration.
4. The method of claim 1 wherein the amount administered reduces glycosaminoglycans (GAG).
5. The method of claim 1 wherein the vector having the AAV9 capsid is administered.
6. The method of claim 1 wherein the vector having AAVrh10 capsid is administered.
7. The method of claim 1 wherein the vector having the AAV8 capsid is administered.
8. The method of claim 1 wherein the rAAV vector is endovascularly administered.
9. The method of claim 1 wherein multiple doses of the rAAV vector are administered.
10. A method to inhibit or treat one or more symptoms of Pompe Disease in a human in need thereof, consisting of:
administering to the human an amount of a recombinant adeno-associated virus (rAAV) vector having an AAV8, AAV9 or AAVrh10 capsid encapsidating a rAAV genome having an open reading frame encoding acid maltase that is effective to inhibit or treat the one or more symptoms of Pompe Disease in the human and an effective amount of an immune suppressant.
11. The method of claim 10 wherein the immune suppressant comprises cyclophosphamide,
a glucocorticoid, cytostatic agents including an alkylating agent, an anti-metabolite, a cytotoxic antibiotic, an antibody, an agent active on immunophilin, a nitrogen mustard, nitrosourea, platinum compound, methotrexate, azathioprine, mercaptopurine, fluorouracil, dactinomycin, an anthracycline, mitomycin C, bleomycin, mithramycin, IL-2 receptor-(CD25-) or CD3-directed antibodies, anti-IL-2 antibodies, ciclosporin, tacrolimus, sirolimus, IFN-beta, IFN-gamma, an opioid, or a TNF-alpha (tumor necrosis factor-alpha) binding agent.
12. The method of claim 10 wherein the rAAV vector and the immune suppressant are co-administered.
13. The method of claim 10 wherein the immune suppressant is administered after the rAAV vector.
14. The method of claim 10 wherein the immune suppressant is administered before the rAAV vector.
15. The method of claim 10 wherein the immune suppressant is systemically administered.
16. The method of claim 10 wherein the vector having the AAV9 capsid is administered.
17. The method of claim 10 wherein the vector having the AAVrh10 capsid is administered.
18. The method of claim 10 wherein the vector having the AAV8 capsid is administered.
19. The method of claim 10 wherein the rAAV vector is endovascularly is administered.
20. A method to inhibit or treat one or more symptoms associated with a deficiency in acid maltase in a human, consisting of: providing a human with a deficiency in acid maltase that is immunotolerized to acid maltase; and administering to the human an amount of a rAAV vector comprising a AAV8, AAV9 or AAVrh10 capsid encapsidating a rAAV genome having an open reading frame encoding acid maltase effective to inhibit or treat the one or more symptoms associated with the deficiency in acid maltase in the human.
21. The method of claim 20 wherein multiple doses of the rAAV vector are administered.
22. The method of claim 20 wherein the vector having the AAV9 capsid is administered.
23. The method of claim 20 wherein the vector having the AAVrh10 capsid is administered.
24. The method of claim 20 wherein the vector having the AAV8 capsid is administered.
25. The method of claim 20 wherein the rAAV vector is endovascularly administered.
26. The method of claim 20 wherein the amount administered reduces glycosaminoglycans (GAG).
US17/118,353 2013-05-15 2020-12-10 Methods to treat pompe disease using a recombinant adeno-associated virus Abandoned US20210085759A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/118,353 US20210085759A1 (en) 2013-05-15 2020-12-10 Methods to treat pompe disease using a recombinant adeno-associated virus

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201361823757P 2013-05-15 2013-05-15
PCT/US2014/038209 WO2014186579A1 (en) 2013-05-15 2014-05-15 Adeno-associated virus mediated gene transfer to the central nervous system
US201514889750A 2015-11-06 2015-11-06
US15/717,450 US20180036388A1 (en) 2013-05-15 2017-09-27 Adeno-associated virus mediated gene transfer to the central nervous system
US15/995,045 US20180271956A1 (en) 2013-05-15 2018-05-31 Methods to treat pompe disease using a recombinant adeno-associated virus
US17/118,353 US20210085759A1 (en) 2013-05-15 2020-12-10 Methods to treat pompe disease using a recombinant adeno-associated virus

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/995,045 Continuation US20180271956A1 (en) 2013-05-15 2018-05-31 Methods to treat pompe disease using a recombinant adeno-associated virus

Publications (1)

Publication Number Publication Date
US20210085759A1 true US20210085759A1 (en) 2021-03-25

Family

ID=51898870

Family Applications (11)

Application Number Title Priority Date Filing Date
US14/889,750 Active US9827295B2 (en) 2013-05-15 2014-05-15 Methods to treat mucopolysaccharide type I or deficiency in alpha-L-iduronidase using a recombinant adeno-associated virus encoding alpha-L-iduronidase
US15/717,450 Abandoned US20180036388A1 (en) 2013-05-15 2017-09-27 Adeno-associated virus mediated gene transfer to the central nervous system
US15/717,358 Pending US20180099030A1 (en) 2013-05-15 2017-09-27 Methods to treat mucopolysaccharide type ii or deficiency in iduronate-2-sulfatase using a recombinant adeno-associated virus encoding iduronate-2-sulfatase
US15/987,490 Abandoned US20180264090A1 (en) 2013-05-15 2018-05-23 Adeno-associated virus mediated gene transfer to the central nervous system
US15/989,405 Abandoned US20180271955A1 (en) 2013-05-15 2018-05-25 Adeno-associated virus mediated gene transfer to the central nervous system
US15/995,045 Abandoned US20180271956A1 (en) 2013-05-15 2018-05-31 Methods to treat pompe disease using a recombinant adeno-associated virus
US15/995,055 Abandoned US20180271957A1 (en) 2013-05-15 2018-05-31 Methods to treat ceroid lipofuscinosis using a recombinant adeno-associated virus
US16/438,143 Abandoned US20190298812A1 (en) 2013-05-15 2019-06-11 Adeno-associated virus mediated gene transfer to the central nervous system
US17/118,395 Abandoned US20210085760A1 (en) 2013-05-15 2020-12-10 Adeno-associated virus mediated gene transfer to the central nervous system
US17/118,353 Abandoned US20210085759A1 (en) 2013-05-15 2020-12-10 Methods to treat pompe disease using a recombinant adeno-associated virus
US17/212,516 Abandoned US20210346473A1 (en) 2013-05-15 2021-03-25 Adeno-associated virus mediated gene transfer to the central nervous system

Family Applications Before (9)

Application Number Title Priority Date Filing Date
US14/889,750 Active US9827295B2 (en) 2013-05-15 2014-05-15 Methods to treat mucopolysaccharide type I or deficiency in alpha-L-iduronidase using a recombinant adeno-associated virus encoding alpha-L-iduronidase
US15/717,450 Abandoned US20180036388A1 (en) 2013-05-15 2017-09-27 Adeno-associated virus mediated gene transfer to the central nervous system
US15/717,358 Pending US20180099030A1 (en) 2013-05-15 2017-09-27 Methods to treat mucopolysaccharide type ii or deficiency in iduronate-2-sulfatase using a recombinant adeno-associated virus encoding iduronate-2-sulfatase
US15/987,490 Abandoned US20180264090A1 (en) 2013-05-15 2018-05-23 Adeno-associated virus mediated gene transfer to the central nervous system
US15/989,405 Abandoned US20180271955A1 (en) 2013-05-15 2018-05-25 Adeno-associated virus mediated gene transfer to the central nervous system
US15/995,045 Abandoned US20180271956A1 (en) 2013-05-15 2018-05-31 Methods to treat pompe disease using a recombinant adeno-associated virus
US15/995,055 Abandoned US20180271957A1 (en) 2013-05-15 2018-05-31 Methods to treat ceroid lipofuscinosis using a recombinant adeno-associated virus
US16/438,143 Abandoned US20190298812A1 (en) 2013-05-15 2019-06-11 Adeno-associated virus mediated gene transfer to the central nervous system
US17/118,395 Abandoned US20210085760A1 (en) 2013-05-15 2020-12-10 Adeno-associated virus mediated gene transfer to the central nervous system

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/212,516 Abandoned US20210346473A1 (en) 2013-05-15 2021-03-25 Adeno-associated virus mediated gene transfer to the central nervous system

Country Status (13)

Country Link
US (11) US9827295B2 (en)
EP (2) EP2996475A4 (en)
JP (4) JP2016523835A (en)
KR (2) KR20220119187A (en)
CN (3) CN115120746A (en)
AU (3) AU2014265417B2 (en)
BR (1) BR112015028605A8 (en)
CA (2) CA2912678C (en)
HK (2) HK1222093A1 (en)
MX (2) MX2015015560A (en)
RU (1) RU2692251C2 (en)
SG (2) SG10201710448SA (en)
WO (1) WO2014186579A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210346473A1 (en) * 2013-05-15 2021-11-11 Regents Of The University Of Minnesota Adeno-associated virus mediated gene transfer to the central nervous system
US11819539B2 (en) 2017-09-22 2023-11-21 The Trustees Of The University Of Pennsylvania Gene therapy for treating Mucopolysaccharidosis type II

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11201507507PA (en) 2013-03-15 2015-10-29 Univ Pennsylvania Compositions and methods for treating mpsi
EP3151866B1 (en) 2014-06-09 2023-03-08 Voyager Therapeutics, Inc. Chimeric capsids
EP3215191A4 (en) 2014-11-05 2018-08-01 Voyager Therapeutics, Inc. Aadc polynucleotides for the treatment of parkinson's disease
CN114717264A (en) 2014-11-14 2022-07-08 沃雅戈治疗公司 Compositions and methods for treating Amyotrophic Lateral Sclerosis (ALS)
JP6863891B2 (en) 2014-11-14 2021-04-21 ボイジャー セラピューティクス インコーポレイテッドVoyager Therapeutics,Inc. Regulatory polynucleotide
US11697825B2 (en) 2014-12-12 2023-07-11 Voyager Therapeutics, Inc. Compositions and methods for the production of scAAV
WO2016115503A1 (en) * 2015-01-16 2016-07-21 Voyager Therapeutics, Inc. Central nervous system targeting polynucleotides
EP3285788A4 (en) 2015-04-23 2018-12-05 University of Massachusetts Modulation of aav vector transgene expression
GB201508025D0 (en) 2015-05-11 2015-06-24 Ucl Business Plc Fabry disease gene therapy
DK3294323T3 (en) * 2015-05-15 2022-01-10 Univ Minnesota ADENO-ASSOCIATED VIRUS FOR THERAPEUTIC SUBMISSION TO THE CENTRAL NERVOUS SYSTEM
US11027024B2 (en) 2015-05-29 2021-06-08 University Of Iowa Research Foundation Methods of delivery of transgenes for treating brain diseases
SG11201803218PA (en) * 2015-10-23 2018-05-30 Univ Iowa Res Found Methods for treating neurodegenerative diseases using gene therapy to delay disease onset and progression while providing cognitive protection
RU2020141860A (en) 2015-11-05 2021-04-01 Дзе Дженерал Хоспитал Корпорейшн INTRATEKAL DELIVERY OF NUCLEIC ACID SEQUENCES CODING ABCD1 FOR ADRENOMYELONEUROPATHY TREATMENT
US20170202931A1 (en) 2016-01-15 2017-07-20 Sangamo Therapeutics, Inc. Methods and compositions for the treatment of neurologic disease
WO2017136500A1 (en) * 2016-02-03 2017-08-10 The Trustees Of The University Of Pennsylvania Gene therapy for treating mucopolysaccharidosis type i
CA3011943A1 (en) * 2016-02-22 2017-08-31 The University Of North Carolina At Chapel Hill Aav-idua vector for treatment of mps i-associated blindness
US11185555B2 (en) 2016-04-11 2021-11-30 Noah James Harrison Method to kill pathogenic microbes in a patient
US11253612B2 (en) 2016-04-15 2022-02-22 The Trustees Of The University Of Pennsylvania Gene therapy for treating mucopolysaccharidosis type II
US11299751B2 (en) 2016-04-29 2022-04-12 Voyager Therapeutics, Inc. Compositions for the treatment of disease
WO2017201258A1 (en) 2016-05-18 2017-11-23 Voyager Therapeutics, Inc. Compositions and methods of treating huntington's disease
BR112018073384A2 (en) 2016-05-18 2019-03-06 Voyager Therapeutics, Inc. modulating polynucleotides
EP3506817A4 (en) 2016-08-30 2020-07-22 The Regents of The University of California Methods for biomedical targeting and delivery and devices and systems for practicing the same
WO2018064098A1 (en) 2016-09-28 2018-04-05 Cohbar, Inc. Therapeutic mots-c related peptides
KR20190109506A (en) * 2017-01-31 2019-09-25 리젠엑스바이오 인크. Treatment of Mucopolysaccharide Type 1 with Fully-Human Glycosylated Human Alpha-L-iduroidase (IDUA)
WO2018156892A1 (en) 2017-02-23 2018-08-30 Adrx, Inc. Peptide inhibitors of transcription factor aggregation
EA201992358A1 (en) 2017-04-03 2020-03-24 Инкоудид Терапьютикс, Инк. TISSELECTIVE EXPRESSION OF TRANSGEN
SG11201909361SA (en) * 2017-04-14 2019-11-28 Regenxbio Inc Treatment of mucopolysaccharidosis ii with recombinant human iduronate-2 sulfatase (ids) produced by human neural or glial cells
US11752181B2 (en) 2017-05-05 2023-09-12 Voyager Therapeutics, Inc. Compositions and methods of treating Huntington's disease
WO2018204786A1 (en) 2017-05-05 2018-11-08 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (als)
AU2018281280A1 (en) 2017-06-07 2020-01-16 Regeneron Pharmaceuticals, Inc. Compositions and methods for internalizing enzymes
US20230137562A1 (en) 2017-06-07 2023-05-04 Adrx, Inc. Tau aggregation inhibitors
JOP20190269A1 (en) 2017-06-15 2019-11-20 Voyager Therapeutics Inc Aadc polynucleotides for the treatment of parkinson's disease
CA3068328A1 (en) 2017-07-06 2019-01-10 The Trustees Of The University Of Pennsylvania Aav9-mediated gene therapy for treating mucopolysaccharidosis type i
JP7229989B2 (en) 2017-07-17 2023-02-28 ボイジャー セラピューティクス インコーポレイテッド Trajectory array guide system
MX2020001187A (en) 2017-08-03 2020-10-05 Voyager Therapeutics Inc Compositions and methods for delivery of aav.
JP7254815B2 (en) 2017-10-03 2023-04-10 プリベイル セラピューティクス,インコーポレーテッド Gene therapy for lysosomal storage diseases
MX2020003965A (en) 2017-10-03 2020-10-05 Prevail Therapeutics Inc Gene therapies for lysosomal disorders.
KR20200078513A (en) 2017-10-03 2020-07-01 프리베일 테라퓨틱스, 인크. Gene therapy for lysosomal disorders
WO2019079240A1 (en) 2017-10-16 2019-04-25 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
US20200237799A1 (en) 2017-10-16 2020-07-30 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
WO2019132624A1 (en) 2017-12-29 2019-07-04 주식회사 헬릭스미스 Adeno-associated virus (aav) vector having hybrid hgf gene introduced thereto
JP2021525245A (en) * 2018-05-22 2021-09-24 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッドThe Brigham and Women’s Hospital, Inc. Gene therapy for Alzheimer's disease
AU2019346447A1 (en) 2018-09-26 2021-04-29 California Institute Of Technology Adeno-associated virus compositions for targeted gene therapy
EP3917944A1 (en) 2019-01-28 2021-12-08 Cohbar Inc. Therapeutic peptides
AU2020272974A1 (en) * 2019-04-10 2021-11-11 Prevail Therapeutics, Inc. Gene therapies for lysosomal disorders
JP2022526021A (en) 2019-04-10 2022-05-20 プリベイル セラピューティクス,インコーポレーテッド Gene therapy for lysosomal disorders
CN114025806A (en) * 2019-04-10 2022-02-08 普利维尔治疗公司 Gene therapy for lysosomal disorders
JP2023517340A (en) * 2020-03-11 2023-04-25 シャンハイ・ビリーフ-デリバリー・バイオメッド・カンパニー・リミテッド Novel use of aspirin compounds in increasing nucleic acid expression
AU2021368725A1 (en) * 2020-10-30 2023-05-25 Immusoft Corporation Methods of administering genetically modified b cells for in vivo delivery of therapeutic agents
EP4284335A1 (en) 2021-02-01 2023-12-06 RegenxBio Inc. Gene therapy for neuronal ceroid lipofuscinoses
EP4288556A1 (en) * 2021-02-05 2023-12-13 Regents of the University of Minnesota Methods for preventing cardiac or skeletal defects in diseases including mucopolysaccharidoses

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10912804B2 (en) * 2007-02-23 2021-02-09 University Of Florida Research Foundation, Incorporated Compositions and methods for treating diseases
US20210346473A1 (en) * 2013-05-15 2021-11-11 Regents Of The University Of Minnesota Adeno-associated virus mediated gene transfer to the central nervous system

Family Cites Families (91)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US200901A (en) * 1878-03-05 Improvement in machines for compressing and purifying air
JPS5929648A (en) * 1982-08-10 1984-02-16 Mitsubishi Chem Ind Ltd Peptide and analgesic agent
DE69031279T2 (en) 1989-12-05 1998-03-19 Ramsey Foundation NEUROLOGICAL ACTIVE SUBSTANCES FOR NASAL ADMINISTRATION TO THE BRAIN
US5624898A (en) 1989-12-05 1997-04-29 Ramsey Foundation Method for administering neurologic agents to the brain
US6407061B1 (en) 1989-12-05 2002-06-18 Chiron Corporation Method for administering insulin-like growth factor to the brain
US5478745A (en) 1992-12-04 1995-12-26 University Of Pittsburgh Recombinant viral vector system
JP3952312B2 (en) 1993-11-09 2007-08-01 メディカル カレッジ オブ オハイオ A stable cell line capable of expressing adeno-associated virus replication genes
PT733103E (en) 1993-11-09 2004-07-30 Targeted Genetics Corp CREATION OF HIGH RECOMBINANT AAV VECTOR TITLES
WO1996017947A1 (en) 1994-12-06 1996-06-13 Targeted Genetics Corporation Packaging cell lines for generation of high titers of recombinant aav vectors
US5656785A (en) 1995-08-07 1997-08-12 The Charles Stark Draper Laboratory, Inc. Micromechanical contact load force sensor for sensing magnitude and distribution of loads and tool employing micromechanical contact load force sensor
US6190659B1 (en) 1996-09-17 2001-02-20 The Rockefeller University Bacterial plasmin binding protein and methods of use thereof
AU5445998A (en) 1996-11-19 1998-06-10 Surgx Corporation A transient voltage protection device and method of making same
JP2001506133A (en) 1996-12-18 2001-05-15 ターゲティッド ジェネティクス コーポレイション AAV split-packaging genes and cell lines containing such genes for use in producing recombinant AAV vectors
AU8672198A (en) * 1997-07-31 1999-02-22 Chiron Corporation Method enabling readministration of aav vector via immunosuppression of host
PT1080202E (en) 1998-05-27 2006-05-31 Avigen Inc DISTRIBUTION OF AAV VECTORS ENCODING AADC INTENSIFIED BY CONVECTION
EP1137401B1 (en) 1998-12-09 2005-11-23 Chiron Corporation Administration of neurotrophic agents to the central nervous system
US7273618B2 (en) 1998-12-09 2007-09-25 Chiron Corporation Method for administering agents to the central nervous system
EP1135105B1 (en) 1998-12-09 2005-03-02 Chiron Corporation Use of a neurologic agent for the manufacture of a medicament for the treatment of a central nervous system disorder
US6585971B1 (en) 1999-11-12 2003-07-01 Harbor-Ucla Research And Education Institute Recombinant α-L-iduronidase, methods for producing and purifying the same and methods for treating disease caused by deficiencies thereof
US6569661B1 (en) 1999-11-12 2003-05-27 Biomarin Pharmaceutical Inc. Recombinant α-L-iduronidase, methods for producing and purifying the same and methods for treating diseases caused by deficiencies thereof
WO2001036603A2 (en) * 1999-11-17 2001-05-25 Avigen, Inc. Recombinant adeno-associated virus virions for the treatment of lysosomal disorders
IL150109A0 (en) 1999-12-09 2002-12-01 Chiron Corp Method for administering a cytokine to the central nervous system and the lymphatic system
US7084126B1 (en) 2000-05-01 2006-08-01 Healthpartners Research Foundation Methods and compositions for enhancing cellular function through protection of tissue components
US20040204379A1 (en) 2000-06-19 2004-10-14 Cheng Seng H. Combination enzyme replacement, gene therapy and small molecule therapy for lysosomal storage diseases
PT1782825E (en) * 2000-07-18 2014-11-04 Univ Duke Treatment of glycogen storage diseases type ii
US20020014242A1 (en) * 2000-07-31 2002-02-07 Abraham Scaria Use of rapamycin to inhibit immune response and induce tolerance to gene therapy vector and encoded transgene products
US20020082215A1 (en) 2000-10-13 2002-06-27 Chiron Corporation Method for treating ischemic events affecting the central nervous system
US20020169102A1 (en) 2001-04-03 2002-11-14 Frey William H. Intranasal delivery of agents for regulating development of implanted cells in the CNS
EP1385937A4 (en) 2001-04-20 2005-11-09 Chiron Corp Delivery of polynucleotide agents to the central nervous sysstem
EP1487474A4 (en) 2002-02-25 2006-11-29 Chiron Corp Intranasal administration of mc4-r agonists
US7485314B2 (en) * 2002-05-06 2009-02-03 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Induction of antigen specific immunologic tolerance
AU2003302724A1 (en) * 2002-08-13 2004-07-09 Mcivor, Scott, R. Methods of using vectors to treat metabolic disorders
CN1703227A (en) * 2002-09-27 2005-11-30 儿童医疗中心有限公司 Methods and compositions for treatment of neurological disorder
US7446098B2 (en) * 2003-02-18 2008-11-04 Mount Sinai School Of Medicine Of New York University Combination therapy for treating protein deficiencies
US20050026823A1 (en) * 2003-06-20 2005-02-03 Biomarin Pharmaceutical Inc. Use of the chaperone receptor-associated protein (RAP) for the delivery of therapeutic compounds to the brain and other tissues
US7442372B2 (en) * 2003-08-29 2008-10-28 Biomarin Pharmaceutical Inc. Delivery of therapeutic compounds to the brain and other tissues
US8071364B2 (en) * 2003-12-24 2011-12-06 Transgenrx, Inc. Gene therapy using transposon-based vectors
US7618615B2 (en) 2004-08-13 2009-11-17 Healthpartners Research Foundation Methods for providing neuroprotection for the animal central nervous system against neurodegeneration caused by ischemia
US7776312B2 (en) 2004-08-13 2010-08-17 Healthpartners Research Foundation Method of treating Alzheimer's disease comprising administering deferoxamine (DFO) to the upper one-third of the nasal cavity
US20090136505A1 (en) 2005-02-23 2009-05-28 Johanna Bentz Intranasal Administration of Active Agents to the Central Nervous System
CA2598666A1 (en) 2005-02-23 2006-08-31 Alza Corporation Intranasal administration of active agents to the central nervous system
JP5276982B2 (en) 2005-08-26 2013-08-28 ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティ Method for the treatment of headache by administration of oxytocin
MX360595B (en) * 2006-02-08 2018-11-09 Genzyme Corp Gene therapy for niemann-pick disease type a.
EP3459441A1 (en) * 2006-02-09 2019-03-27 Genzyme Corporation Slow intraventricular delivery
US8153604B2 (en) 2006-04-24 2012-04-10 Geron Corporation CNS-tumor treatment method and composition
EP1915986A1 (en) 2006-10-23 2008-04-30 BIOPHARM GESELLSCHAFT ZUR BIOTECHNOLOGISCHEN ENTWICKLUNG VON PHARMAKA mbH Lipid growth factor formulations
WO2008103993A2 (en) * 2007-02-23 2008-08-28 University Of Florida Research Foundation, Inc. Compositions and methods for treating glycogen storage diseases
ES2635726T3 (en) * 2007-06-06 2017-10-04 Genzyme Corporation Gene therapy for lysosomal storage diseases
JP2011504163A (en) 2007-06-08 2011-02-03 ヘルスパートナーズ リサーチ ファウンデーション Pharmaceutical compositions and methods for enhancing targeting of therapeutic compounds to the central nervous system
US8283160B2 (en) 2007-09-11 2012-10-09 Frey Ii William H Methods, pharmaceutical compositions and articles of manufacture for administering therapeutic cells to the animal central nervous system
EP2058401A1 (en) * 2007-10-05 2009-05-13 Genethon Widespread gene delivery to motor neurons using peripheral injection of AAV vectors
WO2009075815A1 (en) * 2007-12-07 2009-06-18 Duke University Immunomodulating gene therapy
WO2009120978A2 (en) * 2008-03-27 2009-10-01 The Ohio State University Treatment of metabolic-related disorders using hypothalamic gene transfer of bdnf and compositions therfor
US11219696B2 (en) * 2008-12-19 2022-01-11 Nationwide Children's Hospital Delivery of polynucleotides using recombinant AAV9
WO2010071832A1 (en) * 2008-12-19 2010-06-24 Nationwide Children's Hospital Delivery of polynucleotides across the blood brain barrier using recombinant aav9
US9415121B2 (en) * 2008-12-19 2016-08-16 Nationwide Children's Hospital Delivery of MECP2 polynucleotide using recombinant AAV9
US7989502B2 (en) 2009-02-06 2011-08-02 Sri International Intranasal delivery of modafinil
ES2634118T3 (en) * 2009-02-11 2017-09-26 The University Of North Carolina At Chapel Hill Modified virus vectors and methods to manufacture and use them
US20110070241A1 (en) * 2009-06-30 2011-03-24 Duke University Methods for modulating immune responses to aav gene therapy vectors
US8622993B2 (en) 2009-12-18 2014-01-07 Healthpartners Research Foundation Device and method for delivering therapeutic substances to the maxillary sinus of a patient
DK2561073T3 (en) * 2010-04-23 2016-12-12 Univ Massachusetts Aav vectors targeted to central nervous system 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
RU2012154576A (en) * 2010-06-25 2014-07-27 Шир Хьюман Дженетик Терапис, Инк. METHODS AND COMPOSITIONS FOR DELIVERY TO CNS HEPARAN-N-SULFATASE
RS61683B1 (en) * 2010-06-25 2021-05-31 Shire Human Genetic Therapies Cns delivery of therapeutic agents
KR20230159646A (en) * 2010-06-25 2023-11-21 샤이어 휴먼 지네틱 테라피즈 인크. Methods and compositions for cns delivery of iduronate-2-sulfatase
US9770410B2 (en) * 2010-06-25 2017-09-26 Shire Human Genetic Therapies, Inc. Methods and compositions for CNS delivery of arylsulfatase A
EP3875107A1 (en) * 2010-06-25 2021-09-08 Shire Human Genetic Therapies, Inc. Pharmaceutical formulation comprising a replacement enzyme for a lysosomal enzyme for use in treating lysosomal storage disease intrathecally
WO2012109570A1 (en) * 2011-02-10 2012-08-16 The University Of North Carolina At Chapel Hill Viral vectors with modified transduction profiles and methods of making and using the same
US9849195B2 (en) 2011-03-31 2017-12-26 University Of Iowa Research Foundation Methods and compositions for treating brain diseases
US10196636B2 (en) * 2011-04-21 2019-02-05 Nationwide Children's Hospital, Inc. Recombinant virus products and methods for inhibition of expression of myotilin
ES2702496T3 (en) * 2011-04-21 2019-03-01 Nationwide Childrens Hospital Inc Recombinant virus products and procedures for the inhibition of miotilin expression
US8609088B2 (en) * 2011-05-10 2013-12-17 Regents Of The University Of Minnesota Intranasal delivery of therapeutic enzymes to the central nervous system for the treatment of lysosomal storage diseases
EP2709670A4 (en) * 2011-05-18 2015-01-21 Childrens Hosp Medical Center Targeted delivery of proteins across the blood brain barrier
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
US9469851B2 (en) * 2011-07-25 2016-10-18 Nationwide Children's Hospital, Inc. Recombinant virus products and methods for inhibition of expression of DUX4
KR20140108519A (en) * 2011-10-12 2014-09-11 시나게바 바이오파르마, 코포레이션 Recombinant human naglu protein and uses thereof
US9821149B2 (en) 2011-10-14 2017-11-21 Healthpartners Research & Education Methods for using ultrasound for enhancing efficiency and targeting of intranasal administration of therapeutic compounds to the central nervous system
WO2013081706A1 (en) * 2011-12-02 2013-06-06 Armagen Technologies, Inc. Methods and compositions for increasing arylsulfatase a activity in the cns
CA2863964C (en) * 2012-02-07 2021-10-26 Global Bio Therapeutics Usa, Inc. Compartmentalized method of nucleic acid delivery and compositions and uses thereof
CA2877428A1 (en) * 2012-06-21 2013-12-27 Association Institut De Myologie Widespread gene delivery of gene therapy vectors
CA3086754C (en) * 2012-08-01 2023-06-13 Nationwide Children's Hospital Recombinant adeno-associated virus 9
US20150210771A1 (en) * 2012-09-13 2015-07-30 Ronald G. Crystal Treatment of brain cancers using central nervous system mediated gene transfer of monoclonal antibodies
PT3115372T (en) * 2012-11-27 2019-06-12 Biomarin Pharm Inc Targeted therapeutic lysosomal enzyme fusion proteins and uses thereof
EP2986632B1 (en) * 2013-04-20 2018-09-05 Research Institute at Nationwide Children's Hospital Recombinant adeno-associated virus delivery of exon 2-targeted u7snrna polynucleotide constructs
RU2018128780A (en) * 2013-07-26 2018-12-05 Юниверсити Оф Айова Рисерч Фаундейшн METHODS AND COMPOSITIONS FOR TREATMENT OF BRAIN DISEASES
CA2922500A1 (en) * 2013-08-27 2015-03-05 Research Institute At Nationwide Children's Hospital Products and methods for treatment of amyotrophic lateral sclerosis
AU2014337783B2 (en) * 2013-10-24 2020-07-02 Uniqure Ip B.V. AAV-5 pseudotyped vector for gene therapy for neurological diseases
EP3134113A4 (en) * 2014-04-25 2017-11-29 University of Florida Research Foundation, Inc. Methods of permitting a subject to receive multiple doses of recombinant adeno-associated virus
DK3294323T3 (en) * 2015-05-15 2022-01-10 Univ Minnesota ADENO-ASSOCIATED VIRUS FOR THERAPEUTIC SUBMISSION TO THE CENTRAL NERVOUS SYSTEM
EP3328399B1 (en) * 2015-07-31 2023-12-27 Regents of the University of Minnesota Modified cells and methods of therapy
WO2018093925A1 (en) * 2016-11-15 2018-05-24 Laoharawee Kanut Method for improving neurological function in mpsi and mpsii and other neurological disorders

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10912804B2 (en) * 2007-02-23 2021-02-09 University Of Florida Research Foundation, Incorporated Compositions and methods for treating diseases
US20210346473A1 (en) * 2013-05-15 2021-11-11 Regents Of The University Of Minnesota Adeno-associated virus mediated gene transfer to the central nervous system

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
The factsheet of route of administration from FDA website:www.fda.gov/drugs/data-standards-manual-monographs/route-administration retrieved on 9/1/22. *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210346473A1 (en) * 2013-05-15 2021-11-11 Regents Of The University Of Minnesota Adeno-associated virus mediated gene transfer to the central nervous system
US11819539B2 (en) 2017-09-22 2023-11-21 The Trustees Of The University Of Pennsylvania Gene therapy for treating Mucopolysaccharidosis type II

Also Published As

Publication number Publication date
RU2692251C2 (en) 2019-06-24
US9827295B2 (en) 2017-11-28
US20160120960A1 (en) 2016-05-05
CN115120745A (en) 2022-09-30
HK1222767A1 (en) 2017-07-14
CA3209821A1 (en) 2014-11-20
US20180036388A1 (en) 2018-02-08
SG11201509419QA (en) 2015-12-30
MX2015015560A (en) 2016-06-17
WO2014186579A1 (en) 2014-11-20
HK1222093A1 (en) 2017-06-23
EP2996475A4 (en) 2017-01-11
AU2018253615A1 (en) 2018-11-22
EP2996475A1 (en) 2016-03-23
US20210085760A1 (en) 2021-03-25
US20210346473A1 (en) 2021-11-11
SG10201710448SA (en) 2018-01-30
EP3622821A1 (en) 2020-03-18
US20180264090A1 (en) 2018-09-20
US20190298812A1 (en) 2019-10-03
CA2912678C (en) 2023-10-10
US20180271955A1 (en) 2018-09-27
CA2912678A1 (en) 2014-11-20
BR112015028605A8 (en) 2019-12-24
JP2020073559A (en) 2020-05-14
KR20160010526A (en) 2016-01-27
JP2023089201A (en) 2023-06-27
US20180271956A1 (en) 2018-09-27
US20180271957A1 (en) 2018-09-27
US20180099030A1 (en) 2018-04-12
AU2021200982A1 (en) 2021-03-11
RU2015152546A (en) 2017-06-20
KR20220119187A (en) 2022-08-26
JP2022065134A (en) 2022-04-26
AU2014265417A1 (en) 2015-12-03
JP2016523835A (en) 2016-08-12
BR112015028605A2 (en) 2017-07-25
AU2014265417B2 (en) 2018-07-26
MX2021009634A (en) 2021-09-08
CN105377039A (en) 2016-03-02
CN115120746A (en) 2022-09-30

Similar Documents

Publication Publication Date Title
US20210085759A1 (en) Methods to treat pompe disease using a recombinant adeno-associated virus
US20230226225A1 (en) Adeno-associated virus for therapeutic delivery to central nervous system
RU2801511C1 (en) Adeno-associated virus-mediated gene transfer into the central nervous system
RU2804953C2 (en) Vector based on adeno-associated virus for therapeutic delivery to central nervous system

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

AS Assignment

Owner name: REGENXBIO INC., MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KOZARSKY, KAREN;REEL/FRAME:054936/0104

Effective date: 20170914

Owner name: REGENTS OF THE UNIVERSITY OF MINNESOTA, MINNESOTA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MCIVOR, R. SCOTT;BELUR, LALITHA R.;LOW, WALTER;AND OTHERS;SIGNING DATES FROM 20160302 TO 20170918;REEL/FRAME:054936/0306

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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