WO2022170038A1 - Administration de virus adéno-associé de polynucléotide cln3 - Google Patents

Administration de virus adéno-associé de polynucléotide cln3 Download PDF

Info

Publication number
WO2022170038A1
WO2022170038A1 PCT/US2022/015228 US2022015228W WO2022170038A1 WO 2022170038 A1 WO2022170038 A1 WO 2022170038A1 US 2022015228 W US2022015228 W US 2022015228W WO 2022170038 A1 WO2022170038 A1 WO 2022170038A1
Authority
WO
WIPO (PCT)
Prior art keywords
cln3
scaav9
genome
mice
months
Prior art date
Application number
PCT/US2022/015228
Other languages
English (en)
Inventor
Mitchell Goldman
Original Assignee
Amicus Therapeutics, 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 Amicus Therapeutics, Inc. filed Critical Amicus Therapeutics, Inc.
Publication of WO2022170038A1 publication Critical patent/WO2022170038A1/fr

Links

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
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0306Animal model for genetic diseases
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present disclosure relates to recombinant adeno-associated virus (rAAV) delivery of a ceroid lipofuscinosis neuronal 3 (CLN3) polynucleotide.
  • rAAV adeno-associated virus
  • the disclosure provides rAAV and methods of using the rAAV for CLN3 gene therapy of the neuronal ceroid lipofuscinosis (NCL) or CLN3-Batten Disease.
  • NCLs Neuronal ceroid lipofuscinoses
  • UDRS Unified Batten Disease Rating Scale
  • the Cln3 Aex7/8 mouse model was created in the early 2000s to mimic the most common disease-causing mutation in CLN3-Batten disease patients: an approximately Ikb mutation that eliminates exons 7 and 8 from the CLN3 gene (Cotman et al., Hum Mol Genet. 2002;ll(22):2709-2721; Mole et al., Eur J Paediatr Neurol. 2001;5:7-10).
  • the mutation is found in a homozygous manner in 85% of the patients and as a heterozygous mutation in combination with point mutations on the other allele in an additional 15% of patients.
  • CLN3 Aex7/8 animals accumulated autofluorescent storage material and ATP Synthase subunit C in the nervous system at various time points, and exhibited astrocyte reactivity in the brain starting at 10 months of age. Subsequent studies detailed altered glutamate receptor function in the cerebellum, corresponding with motor deficits on an accelerating rotarod assay (Cotman et al., Hum Mol Genet. 2002;ll(22):2709-2721).
  • Cln3 Aex7/8 mice have been characterized at both young and mature time points, where neurodevelopmental motor delays were seen in neonatal and young adult mice, and deficits in gait and hind limb clasping were seen at 10-12 months of age (Cotman et al., Hum Mol Genet. 2002;ll(22):2709-2721; Osorio et al., Genes Brain Behav. 2009 Apr; 8(3): 337-345).
  • CLN3 Aex7/8 mice do not appear to have functional visual impairments, but present with a slight survival deficit when compared to controls at 12 months of age (Cotman et al., Hum Mol Genet.
  • adeno-associated virus 9 encoding a CLN3 polypeptide, comprising a rAAV9 genome comprising in 5’ to 3’ order: a P546 promoter, and a polynucleotide encoding the CLN3 polypeptide.
  • the rAAV9 genome comprises a self-complementary genome (scAAV9).
  • the P546 promoter comprises the sequence of SEQ ID NO: 3.
  • the rAAV9 genome comprises a single-stranded genome.
  • scAAV9 Self-complementary recombinant adeno-associated virus 9
  • the scAAV9 genome further comprises inverted terminal repeats.
  • the genome of the scAAV9 comprises in 5' to 3' order: a first AAV inverted terminal repeat, a P546 promoter comprising the sequence of SEQ ID NO: 3, a polynucleotide encoding the CLN3 polypeptide set out in SEQ ID NO: 1 and a second AAV inverted terminal repeat.
  • the scAAV9 genome further comprises an SV40 intron.
  • the SV40 intron is located within the scAAV9 genome in 5' to 3' order: a first AAV inverted terminal repeat, a P546 promoter comprising the sequence of SEQ ID NO: 3, an SV40 intron, a polynucleotide encoding the CLN3 polypeptide of SEQ ID NO: 1 and a second AAV inverted terminal repeat.
  • the scAAV9 genome further comprises a bovine growth hormone polyadenylation poly A sequence.
  • the bovine growth hormone polyadenylation poly A sequence is located within the scAAV9 genome in 5' to 3' order: a first AAV inverted terminal repeat, a P546 promoter comprising the sequence of SEQ ID NO: 3, a polynucleotide encoding the CLN3 polypeptide of SEQ ID NO: 1, a bovine growth hormone polyadenylation poly A sequence and a second AAV inverted terminal repeat.
  • the scAAV9 genome comprises a polynucleotide sequence at least 90% identical to SEQ ID NO: 4.
  • the polynucleotide encoding the CLN3 polypeptide is at least 90% identical to SEQ ID NO: 2.
  • the therapeutically effective amount of the composition is capable of reversing the disease progression.
  • the dosage capable of reversing the progression refers to an amount of the composition that is sufficient to palliate, ameliorate, stabilize, reverse or slow the progression of the disease, ameliorate of side effects, or otherwise reduce the pathological or phenotypical consequences of the disease.
  • the methods, medicaments or compositions for treatment result in a subject is capable of reversing the progression of the disease, which includes one or more of the following: (a) reduced or slowed lysosomal accumulation of autofluorescent storage material, (b) reduced or slowed lysosomal accumulation of ATP Synthase Subunit C, (c) reduced or slowed glial activation (astrocytes and/or microglia) activation, (d) reduced or slowed astrocytosis, (e) reduced or slowed brain volume loss measured by MRI, (f) reduced or slowed onset of seizures, and (g) stabilization, reduced or slowed progression, or improvement in one or more of the scales that are used to evaluate progression and/or improvement in CLN3 Batten-disease, e.g., the Unified Batten Disease Rating System (UBDRS) assessment scales or the Hamburg Motor and Language Scale, compared to the subject prior to administering the composition or to an untreated CL
  • the therapeutically effective amount of the composition is in a range of from about 1 x 10 12 to about 1 x 10 15 vg, from about 6 x 10 13 to about 4 x 10 14 vg, from about 1.2 x 10 14 to about 4 x 10 14 vg or from about 2 x 10 14 to about 4 x 10 14 vg of the scAAV9 genome.
  • the therapeutically effective amount of the composition is about 3 x 10 13 vg .
  • the therapeutically effective amount of the composition is about 6 x 10 13 vg.
  • the therapeutically effective amount of the composition is about 3 x 10 14 vg.
  • the compositions, rAAV9, ssAAV9, scAAV9 and/or nucleic acid molecules are administered via a route selected from the group consisting of intrathecal, intracistemal, lumbar puncture, intracranial, intracerebroventricular, intraparenchymal, intravenous and combinations thereof.
  • the intrathecal route comprises the intra cisterna magna (ICM) route.
  • the composition is administered via the intracistemal route.
  • the composition is administred via the lumbar puncture route.
  • the composition is administered via the intracranial route.
  • the composition is delivered to the brain or spinal cord.
  • the composition or medicament is delivered to the brain or spinal cord refers to the composition being delivered to a brain stem, or may be delivered to the cerebellum, or may be delivered to a visual cortex, or may be delivered to a motor cortex.
  • the composition delivered to the brain or spinal cord refers to the composition being delivered to a nerve cell, a glial cell, or both.
  • the composition delivered to the brain or spinal cord comprises delivery to a cell of the nervous system such as a neuron, a lower motor neuron, a microglial cell, an oligodendrocyte, an astrocyte, a Schwann cell, or a combination thereof.
  • a cell of the nervous system such as a neuron, a lower motor neuron, a microglial cell, an oligodendrocyte, an astrocyte, a Schwann cell, or a combination thereof.
  • the composition does not comprise a non-ionic, low-osmolar contrast agent (e.g. iohexol).
  • a non-ionic, low-osmolar contrast agent e.g. iohexol
  • the compositions comprises a non-ionic, low-osmolar contrast agent.
  • the compositions comprise a non-ionic, low-osmolar contrast agent is selected from the group consisting of iobitridol, iohexol, iomeprol, iopamidol, iopentol, iopromide, ioversol, ioxilan, and combinations thereof.
  • the composition can be prepared and administered using a therapeutically effective procedure, including by mixing the scAAV9 genome with the contrast agent prior to delivering to the subject.
  • the scAAV9 genome and the contrasting contrast agent are delivered sequentially.
  • the subject can be held in the Trendelenberg position after administering the rAAV9, ssAAV9, or the scAAV or the nucleic acid molecules disclosed herein.
  • the method further comprising administering the subject myelographic contrast agent for CT-scan assisted intra-cistema magna (ICM) suboccipital administration.
  • the myelographic contrast agent does not comprise a non-ionic, low-osmolar contrast agent.
  • the treatment results in stabilization, reduced progression, or improvement in one or more of: the UBDRS subscale score, cognitive function, neuropsychological function, disease severity, pediatric quality of like and gait.
  • a mean yearly rate of change in UBDRS physical impairment score of the subject is less than that of an untreated subject.
  • Figure 1 provides schematics of the ( Figure 1A) the scAAV9.P546.CLN3 gene cassette and ( Figure IB) plasmid construct pAAV.P546.CLN3.
  • KAN used for production of scAAV9.P546.CLN3.
  • a human CLN3 cDNA was inserted under the control of the P546 promoter between Inverted Terminal Repeat (ITR) structures derived from AAV2.
  • ITR Inverted Terminal Repeat
  • SV40 Intron upstream of human CLN3 cDNA
  • BGH Poly A Bovine Growth Hormone polyadenylation
  • KAN is set out in SEQ ID NO: 5.
  • the genome is packaged in AAV9 capsid protein;
  • Figure 2 provides images showing the presence of human CLN3 transcript in CLN3 Aex7/8 mice injected with scAAV9.P546.CLN3;
  • Figure 3 provides graphs showing early reduction in ASM accumulation at 2 months post injection in CLN3 Aex7/8 mice injected with scAAV9.P546.CLN3. The y-axis on all graphs shown represent total area of ASM accumulation.
  • FIG. 4 provides graphs for CLN3 Aex7/8 mice injected with scAAV9.P546.CLN3-injected CLN3 Aex7/8 mice;
  • A motor cortex
  • B somatosensory cortex
  • C visual cortex
  • D thalamus
  • E hindbrain
  • F cerebellum
  • G spinal cord in PBS-injected wild type mice (“WT”), PBS-injected CLN3 Aex7/8 (“CLN3”), and scAAV9.P546.CLN3-injected CLN3 Aex7/8 (“CLN3-AAV”);
  • Figure 5 provides images and graphs demonstrating that ICV administration of scAAV9.P546.CLN3 reduced the aberrant lysosomal accumulation of the mitochondrial protein ATP Synthase subunit C in the brains of 4 and 6 month old CLN3 Aex7/8 mice. Representative images of frozen tissue sections stained for ATP synthase subunit C and visualized with DAB staining at the 6 month time point are provided in the top panels.
  • the graphs in the lower panels provide the quantification of subunit C accumulation in PBS- injected WT (“WT”), PBS-injected CLN3 Aex7/8 (“CLN3”), and scAAV9.P546.CLN3-injected CLN3 Aex7/8 (“CLN3-AAV”) mice at 4 months (4M) and 6 months (6M) post-injection in the somatosensory 1 barrel field of the cortex (S1BF) at 4 and 6 months post-injection and the ventral posteromedial/ventral posterolateral nucleus (VPM/VPL).
  • N 5, p ⁇ 0.0001 between untreated CLN3 Aex7/8 and scAAV9.P546.CLN3 treated animals, as well as wild type animals. p ⁇ 0.5 between wild type and treated CLN3 Aex7/8 mice in the SIBF at 4 and 6 months postinjection;
  • FIG. 6 provides images and graphs for ICV administration of scAAV9.P546.CLN3 reduced astrocytosis in the brains of 4 and 6 month old CLN3 Aex7/8 mice.
  • Top Representative images (6 month time point) of fixed tissue sections stained for GFAP as a marker for activated astrocytes and visualized with DAB staining.
  • N 5 for each group and time point.
  • SIBF barrel cortex.
  • VPM/VPL ventral posteromedial/ventral posterolateral nucleus;
  • FIG. 7 provides images and graphs for ICV administration of scAAV9.P546.hCLN3 reduced microglia activation in the brains of 4 and 6 month old CLN3 Aex7/8 mice.
  • Top Representative images (6 month ) of fixed tissue sections stained for CD68 as a marker for activated microglia and visualized with DAB staining.
  • N 5 for each group and time point.
  • S1BF barrel cortex.
  • VPM/VPL ventral posteromedial/ventral posterolateral nucleus;
  • Figure 8 provides graphs showing Rotarod analysis of wild type and PBS or treated CLN3 Aex7/8 mice up to 18 months post injection. All mice both genders (top panel), male only (middle panel), female only (bottom panel);
  • Figure 9 provides graphs showing Morris Water Maze performance of wild type and PBS or scAAV9.P546.CLN3 treated CLN3 Aex78 mice at up to 18 months of age. All mice (top panels), male only (middle panels), female only (bottom panels);
  • Figure 10 provides graphs showing performance of scAAV9.P546.CLN3-treated CLN3 Aex7/8 mice and PBS-treated mice in the pole climbing assay.
  • the pole climbing assay in which mice are place face up on a vertical pole and time for them to turn and descend along with number of falls measures balance and agility. All mice (top panels), male only (middle panels), female only (bottom panels);
  • Figure 11 provides graphs showing scAAV9.P546.CLN3-treated CLN3 Aex7/8 mice fall less often from vertical poles compared to PBS-treated CLN3 Aex7/8 mice. Mice are place face up on a vertical pole and number of falls measures while attempting to turn around are measured for balance and agility. All mice (top panel), male only (middle panel), female only (bottom panel);
  • Figure 12 provides images showing immunofluorescent Western Blot detection of GFP protein in various brain regions as well as peripheral mouse tissues three weeks post injection with scAAV9.P546.GFP;
  • Figure 13 provides a graph showing reverse transcription quantitative PCR of expression of human CLN3 in various brain regions of a four- year old Cynomolgus Macaque 12 weeks post intrathecal lumbar injection of 3 x 10 13 vg scAAV9.P546.CLN3. The values were normalized to the level of CLN3 protein in the lumbar spinal cord 4-7 ;
  • Figure 14 provides the nucleic acid sequence of scAAV9.P546.CLN3 gene cassette (SEQ ID NO: 4).
  • the AAV2 ITR nucleic acid sequence is in italics (5’ITR is set out as SEQ ID NO: 6 and the 3’ ITR is set out as SEQ ID NO: 9), the P546 promoter nucleic acid sequence (SEQ ID NO: 3) is underlined with a single line, the SV40 intron nucleic acid sequence (SEQ ID NO: 7) is underlined with a double line, the nucleic acid sequence of the human CLN3 cDNA sequence (SEQ ID NO: 2) is in bold, the nucleic acid sequence of the BGH polyA terminator (SEQ ID NO: 8) is underlined with a dotted line;
  • Figure 15 provides the nucleic acid sequence of full length scAAV.P546.CLN3 (SEQ ID NO: 5);
  • Figure 16 provides data demonstrating that scAAV9.p546.CLN3 treatment results in increased levels of hCLN3 transcript expression in the cerebral cortex and spinal cord of Cln3 ‘ 78 mice as measured by qPCR up to 24 months of age. Mean ⁇ SEM, ordinary one-way ANOVA at each month, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 17 provides images showing that scAAV9.p546.CLN3 treatment produces stable hCLN3 transcript throughout the brain of Cln3 ‘ 78 mice as measured by RNAscope (red fluorescence), up to 24 months of age. Images taken at 20X;
  • Figure 18 provides data demonstrating that scAAV9.p546.CLN3 treatment prevented and reduced ASM accumulation in two areas of the brain in Cln3 ‘ 78 mice up to 24 months of age. Mean ⁇ SEM, ordinary one-way ANOVA at each month, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001. Images taken at 20X;
  • Figure 19 provides data demonstrating that scAAV9.p546.CLN3 treatment prevented large amounts of SubUnitC accumulation, (a constituent of the ASM) in two areas of the brain of Cln3 ‘ 78 mice up to 24 months of age. Mean ⁇ SEM, ordinary one-way ANOVA at each month, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001. Images taken at 20X;
  • Figure 20 provides data demonstrating that scAAV9.p546.CLN3 treatment prevented astrocyte activation (GFAP + ) in two areas of the Cln3 ‘ 78 brain up to 24 months of age. Mean ⁇ SEM, ordinary one-way ANOVA at each month, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001. Images taken at 20X;
  • Figure 21 provides data demonstrating scAAV9.p546.CLN3 treatment prevented some microglial activation (CD68 + ) in the two areas of the Cln3 ‘ 78 brain up to 24 months of age, dependent on time point.
  • Mean ⁇ SEM ordinary one-way ANOVA at each month, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001. Images taken at 20X;
  • Figure 22 provides data demonstrating scAAV9.CB.CLN3 treatment is similarly effective in preventing various Batten disease pathologies in 6 and 12 month old Cln3 ‘ 78 mice.
  • Mean ⁇ SEM ordinary two-way ANOVA, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 23 provides data demonstrating Cln3 ‘ 78 have no red blood cell abnormalities, as measured up to 24 months of age. Mean ⁇ SEM, ordinary two-way ANOVA;
  • Figure 24 provides data C In 3 178 mice have no white blood cell abnormalities, as measured up to 24 months of age. Mean ⁇ SEM, ordinary two-way ANOVA;
  • Figure 25 demonstrates scAAV9.p546.CLN3 treated mice show differing levels of SubC accumulation in the CA3 region of the hippocampus based on sex at 12 months of age.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 26 demonstrates scAAV9.p546.CLN3 mice show subtle, differing levels of SubC accumulation in the Piriform Cortex (PIRC) based on sex at multiple time points.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 27 demonstrates scAAV9.p546.CLN3 mice show differing levels of SubC accumulation in the Reticular Thalamic Nucleus (RTN) based on sex at multiple time points.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 28 demonstrates scAAV9.p546.CLN3 mice show differing levels of SubC accumulation in the Somatosensory Cortex based on sex at 12 months.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 29 demonstrates scAAV9.p546.CLN3 mice show differing levels of SubC accumulation in the VPM/VPL of the thalamus based on sex at 12 months.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 30 demonstrates scAAV9.p546.CLN3 mice show differing levels of SubC accumulation in the Basolateral Amygdala (BLA) based on sex at 12 months. Mean ⁇ SEM, Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 31 provides scAAV9.p546.CLN3 mice show differing levels of SubC accumulation in the Polymorphic Layer of the Dentate Gyrus (DG) based on sex at 12 and 18 months.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 32 demonstrates scAAV9.p546.CLN3 mice show differing levels of SubC accumulation in the Habenula (Hab) based on sex at 12 and 18 months.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 33 provides data demonstrating scAAV9.p546.CLN3 mice show differing levels of SubC accumulation in the Mediodorsal Nucleus (MD) based on sex at 12 and 18 months.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 34 demonstrates that scAAV9.p546.CLN3 mice show no difference in levels of SubC accumulation in the Retrosplenial Cortex (RSC) based on sex.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 35 demonstrates that scAAV9.p546.CLN3 mice show differing levels of activated microglia (CD68 + ) in the Somatosensory Cortex (S1BF) based on sex at 6 months.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 36 demonstrates that scAAV9.p546.CLN3 mice show differing levels of microglia (CD68 + ) activation in the VPM-VPL, thalamus based on sex.
  • Mean ⁇ SEM Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 37 demonstrates that scAAV9.p546.CLN3 mice show differing levels of activated microglia (CD68 + ) in the Mediodorsal Nucleus (MD) based on sex. Mean ⁇ SEM, Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001; [0065] Figure 38 demonstrates that scAAV9.p546.CLN3 mice show differing levels of activated microglia (CD68 + ) in the Submedial Nucleus (SM) based on sex. Mean ⁇ SEM, Ordinary Two-way ANOVA with Tukey's post-hoc test, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001;
  • Figure 39 shows the study design of an open-label, dose-escalation, phase l/2a study that included a low-dose and a high-dose cohort of CLN3 patients.
  • Figures 40A-40D provide the UBDS physical impairment score and UBDS capability with actual vision score over time.
  • Figure 41 shows that the mean (SD) yearly rate of change in UBDRS physical impairment score in the three low-dose subjects was 0.07 (3.33).
  • SD mean
  • JNCL predicted an average increase of 2.86 points per year (95% CI: 2.27-3.45).
  • the present disclosure provides methods and products for treating CLN3-Batten Disease.
  • the methods involve delivery of a CLN3 polynucleotide to a subject using rAAV as a gene delivery vector.
  • Adeno-associated virus is a replication-deficient parvovirus, the singlestranded DNA genome of which is about 4.7 kb in length including 145 nucleotide inverted terminal repeat (ITRs) and may be used to refer to the virus itself or derivatives thereof. The term covers all subtypes and both naturally occurring and recombinant forms, except where specified otherwise.
  • ITRs nucleotide inverted terminal repeat
  • the serotypes of AAV are each associated with a specific clade, the members of which share serologic and functional similarities. Thus, AAVs may also be referred to by the clade.
  • AAV9 sequences are referred to as “clade F” sequences (Gao et al., J.
  • AAV-1 is provided in GenBank Accession No. NC_002077
  • AAV-2 is provided in GenBank Accession No. NC_001401 and Srivastava et al., J. Virol., 45: 555-564 ⁇ 1983
  • the complete genome of AAV-3 is provided in GenBank Accession No. NC_1829
  • the complete genome of AAV-4 is provided in GenBank Accession No.
  • AAV-5 genome is provided in GenBank Accession No. AF085716
  • the complete genome of AAV-6 is provided in GenBank Accession No. NC_00 1862
  • at least portions of AAV-7 and AAV-8 genomes are provided in GenBank Accession Nos. AX753246 and AX753249, respectively
  • the AAV -9 genome is provided in Gao et al., J. Virol., 78: 6381- 6388 (2004)
  • the AAV-10 genome is provided in Mol. Ther., 13(1): 67-76 (2006)
  • the AAV-11 genome is provided in Virology, 330(2): 375-383 (2004)
  • portions of the AAV-12 genome are provided in Genbank Accession No.
  • DQ813647; portions of the AAV-13 genome are provided in Genbank Accession No. EU285562.
  • the sequence of the AAV rh.74 genome is provided in see U.S. Patent 9,434,928, incorporated herein by reference.
  • the sequence of the AAV-B1 genome is provided in Choudhury et al., Mol. Ther., 24( ): 1247-1257 (2016).
  • C/.s-acling sequences directing viral DNA replication (rep), encapsidation/packaging and host cell chromosome integration are contained within the ITRs.
  • Three AAV promoters (named p5, pl9, and p40 for their relative map locations) drive the expression of the two AAV internal open reading frames encoding rep and cap genes.
  • the two rep promoters (p5 and pl 9), coupled with the differential splicing of the single AAV intron (at nucleotides 2107 and 2227), result in the production of four rep proteins (rep 78, rep 68, rep 52, and rep 40) from the rep gene.
  • Rep proteins possess multiple enzymatic properties that are ultimately responsible for replicating the viral genome.
  • the cap gene is expressed from the p40 promoter and it encodes the three capsid proteins VP1, VP2, and VP3.
  • the amino acid sequence of the capsid protein VP1 for AAV9 is provided herein as SEQ ID NO: 10.
  • Alternative splicing and non-consensus translational start sites are responsible for the production of the three related capsid proteins.
  • a single consensus polyadenylation site is located at map position 95 of the AAV genome. The life cycle and genetics of AAV are reviewed in Muzyczka, Current Topics in Microbiology and Immunology, 158: 97-129 (1992).
  • AAV possesses unique features that make it attractive as a vector for delivering foreign DNA to cells, for example, in gene therapy.
  • AAV infection of cells in culture is noncytopathic, and natural infection of humans and other animals is silent and asymptomatic.
  • AAV infects many mammalian cells allowing the possibility of targeting many different tissues in vivo.
  • AAV transduces slowly dividing and non-dividing cells, and can persist essentially for the lifetime of those cells as a transcriptionally active nuclear episome (extrachromosomal element).
  • the native AAV pro viral genome is infectious as cloned DNA in plasmids which makes construction of recombinant genomes feasible.
  • the signals directing AAV replication, genome encapsidation and integration are contained within the ITRs of the AAV genome, some or all of the internal approximately 4.3 kb of the genome (encoding replication and structural capsid proteins, repcap) may be replaced with foreign DNA such as a gene cassette containing a promoter, a DNA of interest and a polyadenylation signal. In some instances, the rep and cap proteins are provided in trans.
  • AAV Another significant feature of AAV is that it is an extremely stable and hearty virus. It easily withstands the conditions used to inactivate adenovirus (56° to 65°C for several hours), making cold preservation of AAV less critical. AAV may even be lyophilized. Finally, AAV-infected cells are not resistant to superinfection.
  • AAV refers to the wild type AAV virus or viral particles.
  • AAV AAV virus
  • AAV viral particle AAV viral particle
  • rAAV refers to a recombinant AAV virus or recombinant infectious, encapsulated viral particles.
  • rAAV rAAV virus
  • rAAV viral particle a recombinant infectious, encapsulated viral particles.
  • rAAV genome refers to a polynucleotide sequence that is derived from a native AAV genome that has been modified. In some embodiments, the rAAV genome has been modified to remove the native cap and rep genes. In some embodiments, the rAAV genome comprises the endogenous 5’ and 3’ inverted terminal repeats (ITRs). In some embodiments, the rAAV genome comprises ITRs from an AAV serotype that is different from the AAV serotype from which the AAV genome was derived.
  • the rAAV genome comprises a transgene of interest (e.g., a CLN3-encoding polynucleotide) flanked on the 5’ and 3’ ends by inverted terminal repeat (ITR).
  • the rAAV genome comprises a “gene cassette.”
  • An exemplary gene cassette is set out in Figure 1A and the nucleic acid sequence of SEQ ID NO: 4.
  • the rAAV genome can be a self- complementary (sc) genome, which is referred to herein as “scAAV genome.”
  • the rAAV genome can be a single-stranded (ss) genome, which is referred to herein as “ssAAV genome.”
  • scAAV refers to an rAAV virus or rAAV viral particle comprising a self- complementary genome.
  • ssAAV refers to an rAAV virus or rAAV viral particle comprising a single-stranded genome.
  • rAAV genomes provided herein may comprise a polynucleotide encoding a CLN3 polypeptide.
  • CLN3 polypeptides comprise the amino acid sequence set out in SEQ ID NO: 1, or a polypeptide with an amino acid sequence that is at least: 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence set forth in SEQ ID NO: 1, and which encodes a polypeptide with CLN3 activity (e.g., at least one of increasing clearance of lysosomal auto-fluorescent storage material, reducing lysosomal accumulation of ATP synthase subunit C, and reducing activation of astrocytes and microglia in a patient when treated as compared to, e.g., the patient prior to treatment).
  • a polypeptide with CLN3 activity e.g., at least one of increasing clearance of lysosomal auto-fluorescent storage material, reducing lysosomal accumulation of ATP synthase subunit C, and reducing activation of astrocytes and microglia in
  • a dosage capable of reversing the progression means an amount that is sufficient to palliate, ameliorate, stabilize, reverse or slow the progression of the disease, ameliorate of side effects, or otherwise reduce the pathological or phenotypical consequences of the disease, including one or more of:
  • rAAV genomes comprise a polynucleotide encoding a CLN3 polypeptide wherein the polynucleotide has the nucleotide sequence set out in SEQ ID NO: 2, or a polynucleotide at least: 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the nucleotide sequence set forth in SEQ ID NO: 2 and encodes a polypeptide with CLN3 activity (e.g., at least one of increasing clearance of lysosomal auto-fluorescent storage material, reducing lysosomal accumulation of ATP synthase subunit C, and reducing activation of astrocytes and microglia in a patient when treated as compared to, e.g.,
  • rAAV genomes provided herein comprise a polynucleotide sequence that encodes a polypeptide with CLN3 activity and that hybridizes under stringent conditions to the nucleic acid sequence of SEQ ID NO: 2, or the complement thereof.
  • stringent is used to refer to conditions that are commonly understood in the art as stringent. Hybridization stringency is principally determined by temperature, ionic strength, and the concentration of denaturing agents such as formamide. Examples of stringent conditions for hybridization and washing include but are not limited to 0.015 M sodium chloride, 0.0015 M sodium citrate at 65-68°C or 0.015 M sodium chloride, 0.0015M sodium citrate, and 50% formamide at 42°C. See, for example, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory (Cold Spring Harbor, N.Y. 1989).
  • the rAAV genomes provided herein comprise one or more AAV ITRs flanking the polynucleotide encoding a CLN3 polypeptide.
  • the CLN3 polynucleotide is operatively linked to transcriptional control elements (including, but not limited to, promoters, enhancers and/or polyadenylation signal sequences) that are functional in target cells to form a gene cassette.
  • transcriptional control elements including, but not limited to, promoters, enhancers and/or polyadenylation signal sequences
  • promoters are the P546 promoter and the chicken P actin promoter.
  • Additional promoters are contemplated herein including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus promoter, as well as human gene promoters such as but not limited to, the actin promoter, the myosin promoter, the elongation factor- la promoter, the hemoglobin promoter, and the creatine kinase promoter.
  • SV40 simian virus 40
  • MMTV mouse mammary tumor virus
  • HSV human immunodeficiency virus
  • LTR long terminal repeat
  • MoMuLV promoter MoMuLV promoter
  • an avian leukemia virus promoter an Epstein-Barr virus immediate early promoter
  • Rous sarcoma virus promoter as
  • a P546 promoter sequence set out in SEQ ID NO: 3 and promoter sequences at least: 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the nucleotide sequence set forth in SEQ ID NO: 3 that are promoters with P546 transcription promoting activity.
  • transcription control elements are tissue specific control elements, for example, promoters that allow expression specifically within neurons or specifically within astrocytes. Examples include neuron specific enolase and glial fibrillary acidic protein promoters.
  • inducible promoters are also contemplated.
  • inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline-regulated promoter.
  • the gene cassette may also include intron sequences to facilitate processing of a CLN3 RNA transcript when expressed in mammalian cells.
  • an intron is the SV40 intron.
  • Packaging refers to a series of intracellular events that result in the assembly and encapsidation of an AAV particle.
  • production refers to the process of producing the rAAV (the infectious, encapsulated rAAV particles) by the packing cells.
  • AAV “rep” and “cap” genes refer to polynucleotide sequences encoding replication and encapsidation proteins, respectively, of adeno-associated virus. AAV rep and cap are referred to herein as AAV “packaging genes.”
  • a “helper virus” for AAV refers to a virus that allows AAV (e.g. wild-type AAV) to be replicated and packaged by a mammalian cell.
  • a variety of such helper viruses for AAV are known in the art, including adenoviruses, herpesviruses and poxviruses such as vaccinia.
  • the adenoviruses may encompass a number of different subgroups, although Adenovirus type 5 of subgroup C is most commonly used.
  • Numerous adenoviruses of human, non-human mammalian and avian origin are known and available from depositories such as the ATCC.
  • Viruses of the herpes family include, for example, herpes simplex viruses (HSV) and Epstein- Barr viruses (EBV), as well as cytomegaloviruses (CMV) and pseudorabies viruses (PRV); which are also available from depositories such as ATCC.
  • HSV herpes simplex viruses
  • EBV Epstein- Barr viruses
  • CMV cytomegaloviruses
  • PRV pseudorabies viruses
  • Helper virus function(s) refers to function(s) encoded in a helper virus genome which allows AAV replication and packaging (in conjunction with other requirements for replication and packaging described herein). As described herein, “helper virus function” may be provided in a number of ways, including by providing helper virus or providing, for example, polynucleotide sequences encoding the requisite function(s) to a producer cell in trans.
  • the rAAV genomes provided herein lack AAV rep and cap DNA.
  • AAV DNA in the rAAV genomes (e.g., ITRs) contemplated herein may be from any AAV serotype suitable for deriving a recombinant virus including, but not limited to, AAV serotypes AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV-10, AAV-11, AAV-12, AAV-13, AAV rh.74 and AAV-B1.
  • AAV serotypes AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV-10, AAV-11, AAV-12, AAV-13, AAV rh.74 and AAV-B1.
  • the nucleotide sequences of the genomes of various AAV serotypes are known in the art.
  • rAAV with capsid mutations are also contemplated. See, for example, Marsic et al., Molecular Therapy, 22(11): 1900-1909 (2014).
  • Modified capsids herein are also contemplated and include capsids having various post- translational modifications such as glycosylation and deamidation. Deamidation of asparagine or glutamine side chains resulting in conversion of asparagine residues to aspartic acid or isoaspartic acid residues, and conversion of glutamine to glutamic acid or isoglutamic acid is contemplated in rAAV capsids provided herein. See, for example, Giles et al., Molecular Therapy, 26(12): 2848-2862 (2016). Modified capsids herein are also contemplated to comprise targeting sequences directing the rAAV to the affected tissues and organs requiring treatment.
  • DNA plasmids provided herein comprise rAAV genomes described herein.
  • the DNA plasmids may be transferred to cells permissible for infection with a helper virus of AAV (e.g., adenovirus, El-deleted adenovirus or herpesvirus) for assembly of the rAAV genome into infectious viral particles with AAV9 capsid proteins.
  • helper virus of AAV e.g., adenovirus, El-deleted adenovirus or herpesvirus
  • Techniques to produce rAAV particles, in which an AAV genome to be packaged, rep and cap genes, and helper virus functions are provided to a cell are standard in the art.
  • rAAV particles require that the following components are present within a single cell (denoted herein as a packaging cell): a rAAV genome, AAV rep and cap genes separate from (i.e., not in) the rAAV genome, and helper virus functions.
  • the AAV rep and cap genes may be from any AAV serotype for which recombinant virus can be derived and may be from a different AAV serotype than the rAAV genome ITRs.
  • Production of pseudotyped rAAV is disclosed in, for example, WO 01/83692, which is incorporated by reference herein in its entirety.
  • AAV capsid proteins may be modified to enhance delivery of the rAAV. Modifications to capsid proteins are generally known in the art. See, for example, US 2005/0053922 and US 2009/0202490, the disclosures of which are incorporated by reference herein in their entirety.
  • a method of generating a packaging cell is to create a cell line that stably expresses all the necessary components for rAAV production.
  • a plasmid (or multiple plasmids) comprising a rAAV genome lacking AAV rep and cap genes, AAV rep and cap genes separate from the rAAV genome, and a selectable marker, such as a neomycin resistance gene, may be integrated into the genome of a cell.
  • rAAV genomes may be introduced into bacterial plasmids by procedures such as GC tailing (Samulski et al., 1982, Proc. Natl. Acad. S6.
  • the packaging cell line may then be infected with a helper virus such as adenovirus.
  • a helper virus such as adenovirus.
  • packaging cells that produce infectious rAAV particles.
  • packaging cells may be stably transformed cancer cells such as HeLa cells, 293 cells and PerC.6 cells (a cognate 293 line).
  • packaging cells may be cells that are not transformed cancer cells such as low passage 293 cells (human fetal kidney cells transformed with El of adenovirus), MRC-5 cells (human fetal fibroblasts), WL38 cells (human fetal fibroblasts), Vero cells (monkey kidney cells) and FRhL-2 cells (rhesus fetal lung cells).
  • rAAV infectious encapsidated rAAV particles
  • the genomes of the rAAV lack AAV rep and cap DNA, that is, there is no AAV rep or cap DNA between the ITRs of the genomes of the rAAV.
  • the rAAV genome can be a self-complementary (sc) genome.
  • a rAAV with a sc genome is referred to herein as a scAAV.
  • the rAAV genome can be a single- stranded (ss) genome.
  • An rAAV with a single-stranded genome is referred to herein as an ssAAV.
  • An exemplary rAAV provided herein is the scAAV named “scAAV9.P546.CLN3.”
  • the scAAV9.P546.CLN3 comprises a human CLN3 cDNA under the control of a truncated Methyl CpG binding protein 2 (MeCP2) promoter herein referred to as the P546 promoter (SEQ ID NO: 3).
  • the scAAV genome also comprises an SV40 Intron (upstream of human CLN3 cDNA) and Bovine Growth Hormone polyadenylation (BGH Poly A) terminator sequence (downstream of human CLN3 cDNA).
  • BGH Poly A Bovine Growth Hormone polyadenylation
  • sequence of this scAAV9.P546.CLN3 gene cassette is set out in SEQ ID NO: 4.
  • the sequence of scAAV9.P546.CLN3 gene cassette comprises a polynucleotide at least: 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the nucleotide sequence set forth in SEQ ID NO: 4.
  • the scAAV genome is packaged in an AAV9 capsid and includes AAV2 ITRs (one ITR upstream of the P546 promoter and the other ITR downstream of the bovine growth hormone (BGH) Poly A terminator sequence).
  • the scAAV9 genome comprises in 5' to 3' order: a P546 promoter comprising the nucleotide sequence of SEQ ID NO: 3, and a polynucleotide encoding the CLN3 polypeptide of SEQ ID NO: 1.
  • the scAAV9 genome further comprises inverted terminal repeats, the inverted terminal repeats are located in 5' to 3' order: a first AAV inverted terminal repeat, a P546 promoter comprising the sequence of SEQ ID NO: 3, a polynucleotide encoding the CLN3 polypeptide of SEQ ID NO: 1 and a second AAV inverted terminal repeat.
  • the scAAV9 genome further comprises an SV40 intron, the SV40 intron is located in 5' to 3' order: a first AAV inverted terminal repeat, a P546 promoter comprising the sequence of SEQ ID NO: 3, an SV40 intron, a polynucleotide encoding the CLN3 polypeptide of SEQ ID NO: 1 and a second AAV inverted terminal repeat.
  • the scAAV9 genome further comprises a bovine growth hormone polyadenylation poly A sequence
  • the bovine growth hormone polyadenylation poly A sequence is located in 5' to 3' order: a first AAV inverted terminal repeat, a P546 promoter comprising the sequence of SEQ ID NO: 3, a polynucleotide encoding the CLN3 polypeptide of SEQ ID NO: 1, a bovine growth hormone polyadenylation poly A sequence and a second AAV inverted terminal repeat.
  • the rAAV may be purified by methods standard in the art, such as by column chromatography or cesium chloride gradients.
  • Methods for purifying rAAV from helper virus are known in the art and may include methods disclosed in, for example, Clark et al., Hum. Gene Ther., 10(6): 1031-1039 (1999); Schenpp and Clark, Methods Mol. Med., 69: 427-443 (2002); U.S. Patent No. 6,566,118 and WO 98/09657.
  • compositions comprising rAAV are also provided.
  • Compositions comprise a rAAV encoding a CLN3 polypeptide.
  • Compositions may include two or more rAAV encoding different polypeptides of interest.
  • the rAAV is scAAV or ssAAV.
  • compositions provided herein comprise rAAV and a pharmaceutically acceptable excipient or excipients.
  • Acceptable excipients are nontoxic to recipients and are preferably inert at the dosages and concentrations employed, and include, but are not limited to, buffers such as phosphate [e.g., phosphate-buffered saline (PBS)], citrate, or other organic acids; antioxidants such as ascorbic acid; low molecular weight polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfact
  • compositions provided herein can comprise a pharmaceutically acceptable aqueous excipient containing a non-ionic, low-osmolar compound such as iobitridol, iohexol, iomeprol, iopamidol, iopentol, iopromide, ioversol, or ioxilan, where the aqueous excipient containing the non-ionic, low-osmolar compound can have one or more of the following characteristics: about 180 mgl/mL, an osmolality by vapor-pressure osmometry of about 322mOsm/kg water, an osmolarity of about 273mOsm/L, an absolute viscosity of about 2.3cp at 20°C and about 1.5cp at 37°C, and a specific gravity of about 1.164 at 37°C.
  • a non-ionic, low-osmolar compound such as iobitridol, iohexol
  • compositions comprise about 20% to about 40% non-ionic, low-osmolar compound or about 25% to about 35% non-ionic, low- osmolar compound.
  • An exemplary composition comprises scAAV or rAAV viral particles formulated in 20mM Tris (pH8.0), ImM MgCh, 200mM NaCl, 0.001% poloxamer 188 and about 20% to about 40% non-ionic, low-osmolar compound.
  • Another exemplary composition comprises scAAV formulated in and IX PBS and 0.001% Pluronic F68. In some embodiments, the composition does not comprise a non-ionic, low-osmolar contrast agent.
  • the composition can be prepared and administered using a therapeutically effective procedure, including by mixing the scAAV9 genome with the contrast agent prior to delivering to the subject.
  • the scAAV9 genome and the contrasting contrast agent are delivered sequentially
  • Dosages of rAAV to be administered in methods of the disclosure will vary depending, for example, on the particular rAAV, the mode of administration, the time of administration, the treatment goal, the individual, and the cell type(s) being targeted, and may be determined by methods standard in the art. Dosages may be expressed in units of viral genomes (vg).
  • Dosages contemplated herein include from about IxlO 11 , about IxlO 12 , about IxlO 13 , about l.lxlO 13 , about 1.2xl0 13 , about 1.3xl0 13 , about 1.5xl0 13 , about 2 xlO 13 , about 2.5 xlO 13 , about 3 x 10 13 , about 3.4 x 10 13 , about 3.5 x 10 13 , about 4x 10 13 , about 4.5x 10 13 , about 5 x 10 13 , about 6xl0 13 , about IxlO 14 , about 1.2 xlO 14 , about 2 xlO 14 , about 3 x 10 14 , about 4x 10 14 , about 5xl0 14 , about IxlO 15 , to about IxlO 16 , or more total viral genomes.
  • Dosages of about IxlO 11 to about IxlO 15 vg, about IxlO 12 to about IxlO 15 vg, about IxlO 12 to about IxlO 14 vg, about IxlO 13 to about 6xl0 14 vg, about 6xl0 13 to about 4xl0 14 vg, about 1.2xl0 14 to about 4xl0 14 vg and about 2xl0 14 vg to about 4xl0 14 vg are also contemplated.
  • One dose exemplified herein is 3xl0 13 vg.
  • Another dose exemplified herein includes 1.2xl0 14 vg.
  • Another dose exemplified herein includes 2 xlO 14 vg. Another dose exemplified herein invcludes 3xl0 14 vg. Another dose exemplified herein invcludes 4xl0 14 vg.
  • Methods of transducing target cells including, but not limited to, cells of the nervous system, nerve or glial cells
  • the cells of the nervous system include neurons, lower motor neurons, microglial cells, oligodendrocytes, astrocytes, Schwann cells or combinations thereof.
  • the term “transduction” is used to refer to the administration/delivery of the CLN3 polynucleotide to a target cell either in vivo or in vitro, via a replication-deficient rAAV of the disclosure resulting in expression of a functional polypeptide by the recipient cell.
  • Transduction of cells with rAAV of the disclosure results in sustained expression of polypeptide or RNA encoded by the rAAV.
  • the present disclosure thus provides methods of administering/delivering to a subject rAAV encoding a CLN3 polypeptide by an intrathecal, intracisternal, lumbar puncture, intracranial, intracerebroventricular, intraparenchymal, intravenous and combinations thereof.
  • Intrathecal delivery refers to delivery into the space under the arachnoid membrane of the brain or spinal cord.
  • intrathecal administration is via intracisternal administration.
  • intrathecal administration is via intra cisterna magna (ICM) administration.
  • the intra cisterna magna (ICM) administration is at the craniocervical junction.
  • the intra cistema magna (ICM) administration is at the suboccipital region.
  • Intrathecal administration is exemplified herein. These methods include transducing target cells (including, but not limited to, nerve and/or glial cells) with one or more rAAV described herein.
  • the rAAV viral particle comprising a polynucleotide encoding a CLN3 polypeptide is administered or delivered to the brain and/or spinal cord of a patient.
  • the polynucleotide is delivered to the brain. Areas of the brain contemplated for delivery include, but are not limited to, the motor cortex, visual cortex, cerebellum, and the brain stem.
  • the polynucleotide is delivered to the spinal cord.
  • the polynucleotide is delivered to a neuron or lower motor neuron. In some embodiments, the polynucleotide is delivered to nerve and glial cells. In some embodiments, the glial cell is a microglial cell, an oligodendrocyte or an astrocyte. In some embodiments, the polynucleotide is delivered to a Schwann cell.
  • the patient is held in the Trendelenberg position (head down position) after administration of the rAAV (e.g., for about 5, about 10, about 15 or about 20 minutes).
  • the patient may be tilted in the headdown position at about 1 degree to about 30 degrees, about 15 to about 30 degrees, about 30 to about 60 degrees, about 60 to about 90 degrees, or about 90 to about 180 degrees).
  • the methods provided herein comprise the step of administering an effective dose, or effective multiple doses, of a composition comprising a rAAV provided herein to a subject (e.g., an animal including, but not limited to, a human patient) in need thereof.
  • a subject e.g., an animal including, but not limited to, a human patient
  • An effective dose is a dose that alleviates (eliminates or reduces) at least one symptom associated with the disease, that slows or prevents progression of the disease, that diminishes the extent of disease, that results in remission (partial or total) of disease, and/or that prolongs survival.
  • methods provided herein result in stabilization, reduced progression, or improvement in one or more of the scales that are used to evaluate progression and/or improvement in CLN3 Batten-disease, e.g. the Unified Batten Disease Rating System (UBDRS) or the Hamburg Motor and Language Scale.
  • UBDRS Unified Batten Disease Rating System
  • the UBDRS assessment scales (as described in Marshall et al., Neurology.
  • methods provided herein may result in one or more of the following: reduced or slowed lysosomal accumulation of autofluorescent storage material, reduced or slowed lysosomal accumulation of ATP Synthase Subunit C, reduced or slowed glial activation (astrocytes and/or microglia) activation; reduced or slowed astrocytosis, and showed a reduction or delay in brain volume loss measured by MRI.
  • the treatment results in stabilization, reduced progression, or improvement in one or more of: the UBDRS subscale score, cognitive function, neuropsychological function, disease severity, pediatric quality of like and gait.
  • a mean yearly rate of change in UBDRS physical impairment score of the subject is less than that of an untreated subject.
  • Combination therapies are also provided.
  • Combination includes either simultaneous treatment or sequential treatment.
  • Combinations of methods described herein with standard medical treatments are specifically contemplated.
  • combinations of compositions e.g., a combination of scAAV9.P546.CLN3 and a contrast agent disclosed herein
  • a combination of scAAV9.P546.CLN3 and a contrast agent disclosed herein for use according to the invention - either simultaneous treatment or sequential treatment - are specifically contemplated.
  • a self-complementary AAV (named scAAV9.P546.CLN3) carrying a CLN3 cDNA under the control of a P546 promoter was produced.
  • the P546 promoter is a truncated version of the MeCP2 promoter, allowing expression of the transgene in both neurons and astrocytes at moderate levels.
  • the efficacy of this gene therapy vector was tested in the CLN3 Aex7/8 knock-in mouse model, which carries the most frequent mutation found in human patients.
  • mice and non-human primates were evaluated in vivo in the CLN3 Aex7/8 knock-in mouse model, wild type mice, and non-human primates and human patient subjects.
  • Data from mice and non-human primates clearly demonstrate efficient transduction of astrocytes and neurons throughout the brain and spinal cord, including deep brain structures, such as thalamus, hippocampus, striatum, amygdala, medulla and cerebellum.
  • a DNA including the open reading frame of human CLN3 (SEQ ID NO: 2) between two Notl restriction sites was synthesized by Eurofin Genomics, USA, and then inserted in a double- stranded AAV2-ITR-based production plasmid.
  • a schematic of the plasmid construct showing the CLN3 DNA inserted between AAV2 ITRs [the 5’ ITR was modified as previously described in McCarty et al., Gene Therapy 8:1248-1254 (2001) to generate scAAV] is shown in Figure 1.
  • the plasmid construct also includes the P546 promoter, an SV40 chimeric intron and a Bovine Growth Hormone (BGH) polyadenylation signal.
  • Figure 14 provides the nucleic acid sequence of scAAV9.P546.CLN3 gene cassette.
  • Figure 15 provides the nucleic acid sequence of full length scAAV.P546.CLN3.
  • scAAV9.P546.CLN3 was produced under cGMP conditions by transient tripleplasmid transfection procedures using the double-stranded AAV2-ITR-based production plasmid, with a plasmid encoding Rep2Cap9 sequence as previously described [Gao et al., J. Virol., 78: 6381-6388 (2004)], along with an adenoviral helper plasmid pHelper (Stratagene, Santa Clara, CA) in HEK293 cells.
  • Virus was purified by two cesium chloride density gradient purification steps, dialyzed against PBS and formulated with 0.001% Pluronic-F68 to prevent virus aggregation and stored at 4°C.
  • scAAV preparations were titered by quantitative PCR using Taq-Man technology. Purity of scAAV was assessed by 4-12% sodium dodecyl sulfate- acrylamide gel electrophoresis and silver staining (Invitrogen, Carlsbad, CA).
  • scAAV9.P546.CLN3 was formulated in 1 x PBS and 0.001% Pluronic F68 or formulated in 20 mM Tris (pH 8.0), 1 mM MgCh, 200 mM NaCl, 0.001% poloxamer 188 and administered into CLN3 Aex7/8 mice via intracerebro ventricular (ICV) injection within 36 hours of birth and expression was monitored at various time points. Wild type and CLN3 Aex7/8 mice injected with an equal volume of PBS served as controls. The effective administered dose was 2.2 x 10 10 vg/mouse using the NCH viral vector core titer.
  • RNAscope in situ hybridization techniques were used to specifically identify human CLN3 mRNA in the brain, cervical spinal cord, thoracic spinal cord and the lumbar spinal cord. This technique involved using RNA in situ hybridization with specific probes to detect only the human transgene encoded by the scAAV9. A strong signal was observed at 4 months and 6 months post- injection, particularly in the cortex (regions A-C) of CLN3 Aex7/8 mice injected with scAAV9.P546.CLN3 compared to no signal in PBS injected controls.
  • ASM autofluorescent storage material
  • ASM autofluorescent storage material
  • scAAV9.P546.CLN3 treated animals displayed minimal signal that was comparable to wild type animals at both, 4 and 6 months post injection (Figure 5) (p ⁇ 0.0001 between PBS and scAAV9.P546.CLN3 treated animals).
  • reactive microglia are primed to release pro-inflammatory mediators such as IL1- /326, which may be a key contributing cause of neuronal cell death at the later stages of CLN3-Batten disease.
  • Activated astrocytes were identified in VPM/VPL thalamus and somatosensory cortex sections by staining for glial fibrillary acidic protein (GFAP) at 4 and 6 month timepoints.
  • GFAP glial fibrillary acidic protein
  • quantification was performed in the barrel cortex within cortical layer IV of the somatosensory cortex. Representative images at 6 months post-injection are shown in Figure 6.
  • Glial activation was also determined in VPM/VPL and somatosensory cortex sections using anti-CD68 staining as a marker for activated microglia.
  • CD68 is a lysosomal protein that is upregulated in cells primed for pro-inflammatory functions such as phagocytosis (Seehafer et al., J Neuroimmunol. 2011;230:169-172). Similar to what was observed with astrocytes, glial activation was significantly reduced in the VPM/VPL and somatosensory cortex regions in the AAV9 injected CLN3 Aex7/8 mice compared to PBS-injected CLN3 Aex7/8 mice after 4 months (Figure 7).
  • mice were subjected to a battery of behavioral testing paradigms including accelerating rotarod assays and pole climbing to test motor function and coordination, as well as Morris water maze to assess learning and memory.
  • behavioral testing paradigms including accelerating rotarod assays and pole climbing to test motor function and coordination, as well as Morris water maze to assess learning and memory.
  • animals have been followed for 10 months post-injection, and studies are ongoing.
  • Previous publications characterizing this mouse model indicate initial delay in neurodevelopmental behavior, followed by normalization and later decline to start at around 10-12 months of age (Osorio et al., Genes Brain Behav. 2009 Apr; 8(3): 337-345).
  • Rotarod analysis showed no statistically significant differences between wild type and PBS or treated CLN3 Aex7/8 mice up to 18 months post-injection.
  • the rotarod assay was performed every 2 months. Mice were placed on an accelerating wheel and time until they fell was measured. At each time point, mice were trained in the morning and testing was performed 4 hours later in the afternoon. Unlike previously published data, no significant difference in performance of wild type mice and PBS CLN3 Aex7/8 mice up to 18 months post- injection was observed (Bosch et al., J Neurosci. 2016; 36(37): 9669-9682). However, a significant difference in latency to fall was observed at 2 months post-injection in female WT mice versus PBS-treated CLN3 Aex7/8 mice. This discrepancy compared to previous data most likely lies in the design of the testing protocol and/or environmental factors in housing.
  • the current protocol used in this study is testing the animals only at one day at each time point, whereas previously published data repeated testing over a time span of 4 days. Moreover, the protocol used in this study was performed at a slightly lower starting speed (36 rpm vs. 40 rpm) and with a longer time interval between morning training and afternoon testing period compared to previously published data (4 hours break vs. 2 hours break). Moreover, the am training set-up was also different: while in the previous study, the mice were trained on a wheel spinning at a constant 5 rpm only in the morning for 5 min, the animals in the current study were trained using the very same settings that were then applied in the afternoon testing, which leads to acceleration of the wheel by 0.3 rpm every 2 seconds.
  • Pole climbing assay showed improved performance of scAAV9.P546.CLN3 treated CLN3 Aex7/8 compared to PBS injected animals.
  • the pole climbing test measures the time the mice take to turn around on a vertical pole when placed on it facing upwards, as well as the time to descend the pole when placed on it facing downwards. Moreover, the number of falls from the pole while attempting to turn or descend is also sometimes measured. This test evaluates coordination and balancing capabilities.
  • the P546 promoter allows expression of transgenes throughout the CNS in a similar manner as the chicken-beta-actin (CBA) promoter.
  • CBA chicken-beta-actin
  • mice were injected with either scAAV9.CB.GFP or scAAV9.P546.GFP formulated in lx PBS and 0.001% Pluronic F68 or 20mM Tris (pH 8.0), 1 mM MgCh, 200 mM NaCl, 0.001% and poloxamer 188 at 5 x 10 10 viral genomes per animal. After 3 weeks, the animals were sacrificed, and the brains were put directly under a fluorescent dissection microscope.
  • Another mouse was injected with scAAV9.P546.GFP and kept alive for 200 days. After 200 days, the animal was sacrificed, and whole-brain sagittal sections were stained for GFP expression. Even 200 days post-injection, widespread expression of the GFP transgene was observed throughout the entire brain, including Cortex, Hippocampus, Midbrain, Medulla, Amygdala, and Cerebellum, further indicating that the P546 promoter is a good candidate for CNS gene therapy.
  • a single dose of 3.4xl0 13 vg scAAV9.P546.CLN3 was formulated in lx PBS and 0.001 % Pluronic F68 and administered into three 3-4 year old cynomolgus macaques.
  • the normalization was performed against the vector-derived CLN3 RNA levels found in the lumbar spinal cord, which was set to 1 rather than to saline-injected or uninjected animals as a normalization to zero was not possible. Actin was used as a normalizing gene.
  • mice were dosed with either PBS, scAAV9.p546.CLN3, or scAAV9.CB.CLN3 gene therapy via intracerebro ventricular (ICV) injection at postnatal day 1.
  • ICV intracerebro ventricular
  • the mice were administered 5xl0 10 vg/animal (4pL volume).
  • the injection method and timing was selected to target specific neuronal populations that are relevant in CLN3-Batten disease patients. Animals were sedated via hypothermia during the procedure, monitored until fully recovered, and genotyped as previously described (see Morgan et al. PLoS One 8; and Laboratory, TJ Protocol 18257: Standard PCR Assay).
  • hCLN3 Forward primer sequence CGCTAGCATCTCATCAGGCCTTG (SEQ ID NO: 11); hCLN3 Reverse primer sequence: AGCATGGACAGCAGGGTCTG (SEQ ID NO: 12).
  • scAAV9.p546.CLN3 treatment resulted in increased levels of hCLN3 transcript expression in the cerebral cortex and spinal cord of Cln3 i7/8 mice as measured by qPCR up to 24 months of age.
  • a single, neonatal, ICV administration of scAAV9.p546.CLN3 results in sustained and well-targeted expression of hCLN3.
  • RNAscope was carried out to detect CLN3 transcript in the brain of the treated mice.
  • Mice were CO2 euthanized and cardiac perfused with PBS. Brains were collected and placed on a 1 mm sagittal brain block. Brains were sliced at the midline and 3 mm right of the midline. The 3 mm sagittal piece was flash frozen with -50°C isopentane and then sectioned on a cryostat at 16 pm and placed on slides. Slides were then processed according to the manufacturer's suggested protocols (ACDBio manuals 320293 and 320513).
  • Sections were labeled with a human specific CLN3 probe (ACDBio Cat No 470241), which consisted of 20 double Z pairs in regions of the CLN3 gene with little homology between mouse and human CLN3 (region 631-1711).
  • Slides were fluorescently labeled with the RNAscope Fluorescent Multiplex Kit (ACDBio Catalog no 320850) using their Amp 4-FL-AltC, which tagged the hCLN3 probe with a 550 nm fluorophore, slides were counterstained with DAPI to label nuclei.
  • Tissue sections were mounted on slides under coverslips using antifade mounting media (Dako Paramount, Agilent). Slides were stored in the dark before imaging. Sections were imaged and analyzed using a Nikon NiE microscope with NIS -Elements Advanced Research software (v4.20).
  • scAAV9.p546.CLN3 treatment produces stable hCLN3 transcript throughout the brain of Cln3 ‘ 78 mice as measured by RNAscope (red fluorescence), up to 24 months of age.
  • the quantitative PCT and the RNAscope assays confirm a single, neonatal, ICV administration of scAAV9.p546. results in sustained and well-targeted expression of hCLN3.
  • scAAV9.p546.CLN3 gene therapy increases hCLN3 gene expression throughout the brain and spinal cord up to 24 months of age.
  • Figure 18 demonstrates that scAAV9.p546.CLN3 treatment prevents and reduces ASM accumulation in two areas of the brain in Cln3 ‘ 78 mice up to 24 months of age.
  • Figure 19 demonstrates that scAAV9.p546.CLN3 treatment generally prevented large amounts of SubUnitC accumulation (a constituent of the ASM) in two areas of the brain of Cln3 l7/ mice up to 24 months of age.
  • Figure 20 demonstrates that scAAV9.p546.CLN3 treatment generally prevents astrocyte activation (GFAP+) in two areas of the Cln3 l7/ brain up to 24 months of age.
  • GFAP+ astrocyte activation
  • Figure 21 demonstrates that scAAV9.p546.CLN3 treatment prevents some microglial activation (CD68+) in the two areas of the Cln3 l7/ brain up to 24 months of age, dependent on time point.
  • scAAV9.p546.CLN3 prevented classic Batten disease pathologies in the brain of Cln3 ‘ 78 mice., including storage material accumulation and glial reactivity.
  • Figure 22 demonstrates that scAAV9.CB.CLN3 treatment is similarly effective in preventing various Batten disease pathologies in 6- and 12-month old Cln3 ‘ 78 mice.
  • CBC red blood cells
  • WBC white blood cell
  • Figure 23 provides data for the following CBCs parameters: RBC count, hemoglobin, hematocrit, mean corpuscular volume, mean corpuscular hemoglobin, mean corpuscular hemoglobin concentration, RBC distribution, platelet count and mean platelet volume.
  • Figure 24 provides data for the following WBCs parameters: WBC count, percent lymphocyte count, percent monocytes, percent granulocytes.
  • scAAV9.p546.CLN3 gene therapy prevents many of the cellular hallmarks of CLN3-Batten disease up to 24 months of age in Cln3 zl7/S mice, including ASM, SubUnit C, GFAP and CD68 expression.
  • scAAV9.CB.CLN3 gene therapy prevents many of the cellular hallmarks of CLN3-Batten disease up to 24 months of age in Cln3 ‘ 78 mice, including ASM, SubUnit C, GFAP and CD68 expression.
  • Wild type (WT) and Cl n3 17/8 mice were dosed with either PBS, scAAV9.p546.CLN3, or scAAV9.CB.CLN3 gene therapy via intracerebro ventricular (ICV) injection at postnatal day 1.
  • the mice were administered 5xl0 10 vg/animal (4pL volume).
  • Wild type and Cln3 ‘ 78 mice were CO2 euthanized, perfused with PBS, and tissue was fixed with 4% PFA. Fixed brains were sectioned on a vibratome at 50 pm (Leica VT10008). Sections were processed with standard immunofluorescence and DAB staining protocols. Primary antibodies included anti-CD68 (AbD Serotec, MCA1957; 1:2000) and anti- ATP synthase subunit C (Abeam, abl81243, 1:1000). Secondary antibodies included anti-rat and anti-rabbit biotinylated (Vector Labs, BA-9400; 1:2000). Sections were imaged and analyzed using an Aperio Slide Scanning Microscope at 20X.
  • Figure 25 demonstrates that mice treated with scAAV9.p546.CLN3 show differing levels of SubC accumulation in the CA3 region of the hippocampus based on sex at 12 months of age. At 12 months of age, treated female Cln3 A7/8 mice accumulated significantly more SubC than wild type, while the accumulation of SubC in treated males did not differ from wild type. However, this difference was not seen at any other time point analyzed.
  • Figure 26 demonstrates mice treated with scAAV9.p546.CLN3 showed subtle, differing levels of SubC accumulation in the Piriform Cortex (PIRC) based on sex at multiple time points.
  • PIRC Piriform Cortex
  • FIG. 27 demonstrates scAAV9.p546.CLN3 treated mice showed differing levels of SubC accumulation in the Reticular Thalamic Nucleus (RTN) based on sex at multiple time points.
  • RTN Reticular Thalamic Nucleus
  • treated female mutant CLN3 mice accumulated significantly more SubC than wild type, while the accumulation of SubC in treated males does not differ from wild type.
  • treated males remained at wild type levels, while treated females have significantly more SubC than both wild type and untreated mutant mice. This difference between female treated and untreated mutants was not present at 18 months, where SubC was significantly higher than WT but significantly lower than untreated mutants in both males and females.
  • Figure 28 demonstrates that scAAV9.p546.CLN3 treated mice show differing levels of SubC accumulation in the Somatosensory Cortex based on sex at 12 months. At 12 months of age, treatment with AAV did not reduce SubC accumulation compared to untreated mutant females, while accumulation of SubC in treated males was prevented and did not differ from wild type. However, this difference was not seen at any other time point analyzed.
  • Figure 29 demonstrates mice treated with scAAV9.p546.CLN3 showed differing levels of SubC accumulation in the VPM/VPL of the thalamus based on sex at 12 months. At 6 months of age, treated females accumulated significantly less SubC than untreated mutant females, but significantly more than wild type females. This difference continued through 12 and 18 months, with no differences between wild type and treated males at any time point analyzed.
  • Figure 30 demonstrates mice treated with scAAV9.p546.CLN3 show differing levels of SubC accumulation in the Basolateral Amygdala (BLA) based on sex at 12 months. At 12 months of age, AAV did not significantly prevent accumulation of SubC in treated female animals. At 18 months, SubC accumulation in treated females remained significantly higher than wild type but was lower than untreated females. By 18 months, treated males also begin to have significantly more SubC than their wild type counterpart, mimicking what was seen in female groups.
  • BLA Basolateral Amygdala
  • Figure 31 demonstrates scAAV9.p546.CLN3 treated mice showed differing levels of SubC accumulation in the Polymorphic Layer of the Dentate Gyrus (DG)based on sex at 12 and 18 months.
  • DG Dentate Gyrus
  • treated females appeared to have accumulated significantly more SubC than both wild type females and untreated mutant females.
  • Raw images revealed a darkened granule cell layer surrounding the polymorphic layer of the dentate gyrus that may impact the thresholding results. This darkening was present only in this group and only at the 12-month time point. This increase was also no longer seen at 18 months in females. However, at 18 months, treated males began to have more SubC accumulation than wild type males.
  • Figure 32 demonstrates scAAV9.p546.CLN3 treated mice show differing levels of
  • Figure 33 demonstrates mice treated with scAAV9.p546.CLN3 showed differing levels of SubC accumulation in the Mediodorsal Nucleus based on sex at 12 and 18 months. At 12 months of age, treated females accumulated significantly less SubC than untreated mutant females, but significantly more than wild type females. This difference was also seen at 18 months, with no differences between wild type and treated males at any time point analyzed.
  • Figure 34 demonstrates mice treated with scAAV9.p546.CLN3 showed no difference in levels of SubC accumulation in the Retrosplenial Cortex (RSC) based on sex. There were no differences between wild type and treated males at any time point analyzed.
  • RSC Retrosplenial Cortex
  • Figure 35 demonstrates mice treated with scAAV9.p546.CLN3 showed differing levels of activated microglia (CD68 + ) in the Somatosensory Cortex (S1BF) based on sex at 6 months.
  • treated females had increased microglial activation compared to wild type and untreated females, with treated males higher than wild type but lower than untreated. No sex differences were seen at 12 and 18 months of age.
  • Figure 36 demonstrates mice treated with scAAV9.p546.CLN3 show differing levels of microglia activation in the VPM-VPL, thalamus based on sex.
  • treated females have increased microglial activation compared to wild type and untreated females, with treated males higher than wild type but lower than untreated. This difference is also seen at 12 months. The same is seen in female groups at 18 months. However, treated males did not significantly differ from wild type at this time point.
  • Figure 37 demonstrates scAAV9.p546.CLN3 mice show differing levels of activated microglia (CD68 + ) in the Mediodorsal Nucleus (MD) based on sex.
  • treated females had increased microglial activation compared to wild type and untreated females, with treated males higher than wild type but not different than untreated males.
  • treated males had higher microglial activation than wild type and lower activation than untreated males, while treatment appeared to have no effect on females.
  • FIG. 38 demonstrates mice treated with scAAV9.p546.CLN3 showed differing levels of activated microglia in the Submedial Nucleus (SM) based on sex.
  • SM Submedial Nucleus
  • the treated animals showed high potency in preventing the accumulation of ceroid lipofuscin in the brain compared to untreated animals. Based on the established target engagement the dose 2.5 x 10 10 vg was determined as the minimum efficacious dose (MED).
  • MED minimum efficacious dose
  • a non-human primate (cynomolgus monkey) study was conducted over 26 weeks for safety and biodistribution. The animals were treated with 1 x 10 13 , 3 x 10 13 or 6 x 10 13 vg of scAAV9.P546.CLN3 per animal. All dose levels were well tolerated without significant sequelae. Based on the results of the study, the non-observed-adverse-effect-level (NOAEL) was identified to be 6 x 10 13 vg/animal, following a single administration via lumber puncture (LP) or intra-cisterna magna (ICM) in cynomolgus monkeys, and hence a margin of exposure (MOE) could be established.
  • NOAEL non-observed-adverse-effect-level
  • the dosing regimen for human subjects can be scaled between species based on average brain mass and an approximation of the volume of the cerebrospinal fluid (CSF).
  • Approximate brain mass for a juvenile-to-adult mouse is 0.4-0.5 g
  • for a cynomolgus nonhuman primate is 70 g
  • for human subjects aged 4 years and older is 1200 g (Kandel et al 2000; Rengachary and Ellenbogen 2005; Dekaban and Sadowsky 2000).
  • CSF cerebral spinal fluid
  • subjects aged 4-years old is 125 mL
  • subjects 5 years and older have CSF volume of 150 mL, which is similar to that of an adult.
  • An approximate CSF volume of a cynomolgus NHP is about 11-12 mF (SD: 1.1 -1.6) (Kazuhito et al 2014; Ballesteros 2020). Accordingly, the human dose can be of 3 x 10 14 vg, which translates to a 3-fold MOE.
  • Table 1 Dose-Regimen: Extrapolation from Mice and NHPs to Human Subjects a Route of administration: ICV; b Non- Primate study: Route of Administration: LP or ICM; c 1200 g refers to brain mass in human
  • Example 8 An open-label, phase l/2a, AAV9-CLN3 gene transfer clinical trial for juvenile neuronal ceroid lipofuscinosis
  • Safety assessments included dose-limiting toxicity, adverse events (AEs), vital signs, physical and neurological exams, blood and urine laboratory parameters, electrocardiogram, immunological assessments and viral shedding.
  • UDRS Unified Batten Disease Rating Scale
  • PEDSQOL Pediatric Quality of Life
  • the UBDRS was developed specifically for monitoring disease progression of JNCL, which includes physical, behavioral, seizure and capability subscales.
  • the primary efficacy assessment for this study is the UBDRS physical subscale (Table 1), which was performed at baseline, Day 30, and Months 3, 6, 9, 12, 18, 24, 30, and 36. The results are reported as descriptive data and no statistical analysis was conducted for this analysis.
  • the scAAV9.P546.CLN3 was formulated in 20 mM Tris (pH8.0), 1 mM MgCh, 200 mM NaCl and 0.001% poloxamerl88.
  • scAAV9.p546.CLN3 containing formulation was administered one-time through an intrathecal catheter inserted by a lumbar puncture into the interspinous into the subarachnoid space of the lumbar thecal sac.
  • additional subject(s) were enrolled as safety concerns were not found after the last subject from the first cohort was evaluated at one-month post-injection.
  • Efficacy data will be compared to available natural history data from juvenile CLN3 Batten patients. At the time of screening, participants had UBDRS score of ⁇ 7 and were able to walk independently for >50 ft.
  • Figures 40A-40C provide the UBDS Physical Impairment score and UBDS capability with actual vision score over time for subjects treated with a low dose of vector genome (6 x 10 13 vg). UBDRS Physical Impairment scores have been recorded up to Month 12 or Month 15 for the 3 low-dose patients.
  • Figure 4D provides the UBDS Physical Impairment score and UBDS capability with actual vision score over time for subjects treated with a high dose of vector genome (IxlO 14 vg). The only high-dose patient had UBDRS evaluations up to Month 3.
  • Figure 41 shows the interim clinical data analysis based on Figures 40A-40D.
  • the mean (SD) yearly rate of change in UBDRS Physical Impairment score in the 3 low-dose subjects was 0.07 (3.33) ( Figure 41).
  • a historical natural history study of 82 patients with JNCL predicted an average increase of 2.86 points per year (95% CI: 2.27-3.45) (see Kwon JM et al. Neurology. 2011;77(20): 1801- 1807). Sample size was not determined through statistical justification.
  • An open-label, dose-escalation (2-cohort) study may be designed to evaluate safety and potential efficacy of a formulation comprising a vector genome-delivered gene transfer of CLN3 in subjects diagnosed with CLN3 Batten disease.
  • the subject may be >4 years old as compared to untreated subjects from a natural history cohort.
  • a primary efficacy of the vector genome treatment may be measured using the Unified Batten Disease Rating Scale (UBDRS) physical subscale score which measures disease progression across 21 items, including motor, strength, gait, balance, speech, and visual acuity. In each category, scores range from 0 (considered normal or having no impairment) to 4 (considered fully impaired or unable to function).
  • UDRS Unified Batten Disease Rating Scale
  • the baseline UBDRS physical subscale score for each of the subjects may be equal to 10 or more to less than or equal to 40.
  • the subject may be able to walk independently at least 5 meters or more.
  • the subject may not have major comorbidities of cardiovascular disease, respiratory disease, metabolic diseases, vertebral anatomical deformities of cervical and lumbar regions of the spinal cord, acute and severe infections.
  • the subject may not have other forms of batter disease and neurodegenerative diseases.
  • the subject may not have received biologic and/or non-biologic investigational products in the past.
  • the patient may not have any contraindications to the ICM suboccipital injection and/or lumbar puncture.
  • Secondary outcomes may evaluate the change from baseline to 36 months for cognitive function (WPPSI-IV, WISC-V, WAIS IV), neuropsychological function (Vineland 3, CBCL, ABCL), disease severity (CLN3SS, brain MRI volumetric measurements), pediatric quality of life (PedsQL, EQ-5D-5L, EQ-5D-Y), and gait (ZenoTM Walkway).
  • the primary analysis for efficacy may be assessed when all patients have completed the three-year study. Basis of determining efficacy may be by stabilization or reduced progression of the disease based on the well-established Unified Batten Disease Rating Scale (UBDRS) that was developed specifically for CLN3-Batten Disease.
  • UBDRS Unified Batten Disease Rating Scale
  • the primary efficacy endpoint of rate of decline in the UBDRS physical subscale score at 36 months may be compared between vector genome-treated subjects and untreated subjects from the natural history cohort. Upon completion of the three-year study period patients may be monitored annually for 5 years as per FDA guidance.
  • the vector genome may comprise a self-complementary recombinant adeno- associated virus 9 (scAAV9), which has been genetically modified to express the human CLN3 gene.
  • the vector genome may contain only the minimal elements required for the expression of the CLN3 transgene, including AAV2 inverted terminal repeats, the P546 promoter (a truncated version of the MECP2 promoter), the human CLN3 cDNA, and an SV40 Poly A signal.
  • the vector genome comprise a sequence encoding the CLN3 polypeptide of SEQ ID NO: 1, wherein the genome of the scAAV9 comprises in 5' to 3' order: a first AAV inverted terminal repeat, a P546 promoter comprising the sequence of SEQ ID NO: 3, a polynucleotide encoding the CLN3 polypeptide of SEQ ID NO: 1 and a second AAV inverted terminal repeat.
  • the formulation may comprise 20 mM Tris (pH 8.0), 1 mM magnesium chloride (MgCh), and 200 mM sodium chloride (NaCl) containing 0.005% poloxamer 188.
  • the vector genome may be supplied in a vial having 0.25 mL formulation with a nominal concentration from 2.0 x 10 13 vg/mL to > 6.0 x 10 13 vg/mL.
  • the vector genome may be supplied in a vial having 0.25 mL formulation with a nominal concentration of > 6.0 x 10 13 vg/mL.
  • the study may comprise administering participants with a single one-time injection of 3.0 x 10 14 vector genomes (vg) in 5mL formulation by an intra-cisterna magna (ICM) suboccipital injection.
  • ICM intra-cisterna magna
  • Gene transfer mediated by the vector genome may provide cells with the corrected CLN3 gene, leading to the production of functional CLN3 protein and prevention of the abnormal accumulation of ceroid lipofuscin and adenosine triphosphate (ATP) synthase subunit C throughout the CNS. This may also reduce abnormal astrocyte and glial activation and may ameliorate the neurological and somatic manifestations of the disease.
  • ATP adenosine triphosphate
  • the subject may be administered systemic corticosteroids at 1 mg/kg of body weight to 2 mg/kg of body weight per day for a total of 30 days.
  • the subject may be administered systemic corticosteroids at a dose of 10 mg to 20 mg per day for a total of 30 days.
  • the subject may be administered systemic corticosteroids at 1 mg/kg of body weight per day for a total of 30 days.
  • subject s vaccination schedule to accommodate concomitant glucocorticoid administration prior to and following vector genome injection.
  • Certain vaccines such as MMR and varicella, may be contraindicated for subjects on a substantially immunosuppressive glucocorticoid dose (i.e., > 2 weeks of daily receipt of 20 mg or 2 mg/kg body weight of prednisone or equivalent).
  • Seasonal respiratory syncytial virus (RSV) prophylaxis may not be precluded.
  • the vector genome administration may elevate transaminase, which may be serious. Specifically, subjects with pre-existing liver impairment or acute hepatic viral infection may be at higher risk of liver injury.
  • the liver function test may be performed at baseline and at the end of the 30-day period of systemic corticosteroid treatment, by clinical examination and by laboratory testing (AST, ALT, total bilirubin, prothrombin time).
  • the liver function test may be performed weekly for the first month and every other week for the second and third months until the results are unremarkable (normal clinical exam, total bilirubin, and prothrombin results, and ALT and AST levels below 2 x ULN).
  • Platelet count blood test may also be performed at baseline and then weekly for the first month, followed by every other week for the second and third months, until platelet counts return to baseline.
  • the vector genome containing vial may be slightly opaque, colorless to faint white solution.
  • the vial may be stored in the freezer at ⁇ -60°C until time to use.
  • the vial may be thawed for 30 minutes to bring to room temperature.
  • the vial may be inspected for particulate matter and/or discoloration prior to infusion.
  • the subject Before administering the vector genome, the subject may be optionally administered with myelographic contrast agent for CT-scan assisted intra-cistema magna (ICM) suboccipital delivery of the vector genome.
  • the myelographic contrast agent may comprise a non-ionic, low-osmolar contrast agent.
  • the myelographic contrast agent may not comprise the non-ionic, low-osmolar contrast agent.
  • the subject may be placed in the lateral decubitus position and administered general anesthesia under standard of care.
  • a spinal needle may be percutaneously inserted into the subarachnoid space between L3-L5 of the spinal vertebral column. Approximately 5 mL of CSF may be withdrawn into a polypropylene tube with a screw top.
  • the withdrawn CSF may not be returned.
  • myelographic contrast agent may be injected into the subarachnoid space.
  • the spine needle may be inserted through an image-guided lateral or midline percutaneous suboccipital injection (above the posterior spinal arch of the Cl cervical vertebra) by advancing the spinal needle into the cistema magna until 5mL clear CSF is obtained.
  • the vector genome may be delivered to the intra-cistema magna (ICM) suboccipital region through the spine needle.
  • the vector genome may be delivered at a rate of approximately 1 mL per 1 minute into the intra-cisterna magna (ICM) suboccipital region.
  • Certain neuroleptics may be contraindicated for use with imaging contrast agents.
  • Discontinuance of neuroleptic drugs including phenothiazines e.g., chlorpromazine, prochlorperazine, and promethazine
  • phenothiazines e.g., chlorpromazine, prochlorperazine, and promethazine
  • the subject may react differently to the treatment if the subject has developed anti- AAV9 antibody after natural exposure prior to or during the treatment.
  • the detection of antibody formation may be highly dependent on the sensitivity and specificity of the assay. Additionally, the incidence of antibody (including neutralizing antibody) positivity in an assay may be influenced by several factors including assay methodology, sample handling, timing of sample collection, concomitant medications, and underlying disease. Accordingly, subjects may be assessed for total anti-AAV9 and anti-CLN3 antibodies by ELISA and IFN-y- secreting. T-cell responses to AAV9 and CLN3 peptides may be measured by ELISpot.
  • the immunogenicity assessments may be done on Day -30 to Day -2 (screening/baseline), at follow-up site visits on Days 7, 14, 21 and 30 (Visits 3 to 6), and at Months 6, 9, 12, 18, and 24 (Visits 7 to 10, Visit 12, and Visit 14).
  • the subject may be monitored for new or worsening cardiac abnormalities that may indicate changes in structure or function, including but not limited to arrhythmias such as tachycardia or bradycardia, sick sinus syndrome, left ventricular hypertrophy, and conduction abnormalities.
  • arrhythmias such as tachycardia or bradycardia
  • sick sinus syndrome such as sick sinus syndrome
  • left ventricular hypertrophy such as conduction abnormalities.
  • Vector shedding after ICM injection with the vector genome may be investigated at multiple time points. Specifically, samples of blood, saliva, urine and stool may be collected at baseline, the day after ICM administration (Day 1), Day 3, Day 7, weekly through Day 30, and monthly through Month 6 and every 3 months thereafter. Sample collection and testing may be stopped when at least 3 consecutive negative samples show negative results.
  • the subject may not be given immunomodulatory treatment concurrently for treating active infections, either acute (such as acute respiratory infections or acute hepatitis) or uncontrolled chronic (such as chronic active hepatitis B).
  • active infections either acute (such as acute respiratory infections or acute hepatitis) or uncontrolled chronic (such as chronic active hepatitis B).

Abstract

La présente divulgation concerne l'administration de virus adéno-associé recombinant (rAAV) d'un polynucléotide de céroïde-lipofuscinose neuronale (CLN3). La divulgation fournit des rAAV et des méthodes d'utilisation du rAAV pour la thérapie génique par CLN3 de la céroïde-lipofuscinose neuronale ou de la maladie de Batten liée à CLN3.
PCT/US2022/015228 2021-02-05 2022-02-04 Administration de virus adéno-associé de polynucléotide cln3 WO2022170038A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202163146110P 2021-02-05 2021-02-05
US63/146,110 2021-02-05
US202163248756P 2021-09-27 2021-09-27
US63/248,756 2021-09-27

Publications (1)

Publication Number Publication Date
WO2022170038A1 true WO2022170038A1 (fr) 2022-08-11

Family

ID=80928779

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/015228 WO2022170038A1 (fr) 2021-02-05 2022-02-04 Administration de virus adéno-associé de polynucléotide cln3

Country Status (2)

Country Link
TW (1) TW202246513A (fr)
WO (1) WO2022170038A1 (fr)

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
WO1995013365A1 (fr) 1993-11-09 1995-05-18 Targeted Genetics Corporation Production de titres eleves de vecteurs d'aav recombinants
WO1995013392A1 (fr) 1993-11-09 1995-05-18 Medical College Of Ohio Lignees cellulaires stables aptes a exprimer le gene de replication du virus adeno-associe
WO1996017947A1 (fr) 1994-12-06 1996-06-13 Targeted Genetics Corporation Lignees cellulaires d'encapsidation utilisees pour la generation de titres hauts de vecteurs aav recombinants
WO1997006243A1 (fr) 1995-08-10 1997-02-20 Pasteur Merieux Serums Et Vaccins Procede de purification de virus par chromatographie
WO1997008298A1 (fr) 1995-08-30 1997-03-06 Genzyme Corporation Purification d'adenovirus et de virus adeno-associe (aav) par voie chromatographique
WO1997009441A2 (fr) 1995-09-08 1997-03-13 Genzyme Corporation Vecteurs aav ameliores pour la therapie genique
WO1997021825A1 (fr) 1995-12-15 1997-06-19 Systemix, Inc. Procede de production de lignees de cellules d'encapsidation retrovirales generant un surnageant retroviral a efficacite de transduction elevee
WO1998009657A2 (fr) 1996-09-06 1998-03-12 Trustees Of The University Of Pennsylvania Methode de therapie genique basee sur des virus adeno-associes de recombinaison
US5786211A (en) 1994-06-06 1998-07-28 Children's Hospital, Inc. Adeno-associated virus materials and methods
US5871982A (en) 1994-10-28 1999-02-16 The Trustees Of The University Of Pennsylvania Hybrid adenovirus-AAV virus and methods of use thereof
WO1999011764A2 (fr) 1997-09-05 1999-03-11 Targeted Genetics Corporation Procedes de generation de preparations de vecteurs de aav recombinants dont le titre est eleve et qui sont exemptes de virus assistant
US6258595B1 (en) 1999-03-18 2001-07-10 The Trustees Of The University Of Pennsylvania Compositions and methods for helper-free production of recombinant adeno-associated viruses
WO2001083692A2 (fr) 2000-04-28 2001-11-08 The Trustees Of The University Of Pennsylvania Vecteurs aav recombinants dotes de capsides aav5 et vecteurs aav5 pseudotypes dans des capsides heterologues
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US20050053922A1 (en) 2003-06-30 2005-03-10 Schaffer David V. Mutant adeno-associated virus virions and methods of use thereof
US20090202490A1 (en) 2003-06-30 2009-08-13 Schaffer David V Mutant adeno-associated virus virions and methods of use thereof
US9434928B2 (en) 2011-11-23 2016-09-06 Nationwide Children's Hospital, Inc. Recombinant adeno-associated virus delivery of alpha-sarcoglycan polynucleotides
US9614423B2 (en) 2012-04-07 2017-04-04 Traugott Weller Method for producing rotating electrical machines
US9613872B2 (en) 2014-09-29 2017-04-04 Kabushiki Kaisha Toshiba Method of manufacturing semiconductor device
US9620777B2 (en) 2013-09-30 2017-04-11 Tdk Corporation Positive electrode and lithium ion secondary battery using thereof
US9818600B2 (en) 2014-03-21 2017-11-14 Hitachi Kokusai Electric, Inc. Substrate processing apparatus and method of manufacturing semiconductor device
WO2020163300A1 (fr) * 2019-02-04 2020-08-13 Research Institute At Nationwide Children's Hospital Administration de virus adéno-associé de polynucléotide cln3

Patent Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
WO1995013365A1 (fr) 1993-11-09 1995-05-18 Targeted Genetics Corporation Production de titres eleves de vecteurs d'aav recombinants
WO1995013392A1 (fr) 1993-11-09 1995-05-18 Medical College Of Ohio Lignees cellulaires stables aptes a exprimer le gene de replication du virus adeno-associe
US5658776A (en) 1993-11-09 1997-08-19 Targeted Genetics Corporation Generation of high titers of recombinant AAV vectors
US5786211A (en) 1994-06-06 1998-07-28 Children's Hospital, Inc. Adeno-associated virus materials and methods
US5871982A (en) 1994-10-28 1999-02-16 The Trustees Of The University Of Pennsylvania Hybrid adenovirus-AAV virus and methods of use thereof
WO1996017947A1 (fr) 1994-12-06 1996-06-13 Targeted Genetics Corporation Lignees cellulaires d'encapsidation utilisees pour la generation de titres hauts de vecteurs aav recombinants
WO1997006243A1 (fr) 1995-08-10 1997-02-20 Pasteur Merieux Serums Et Vaccins Procede de purification de virus par chromatographie
WO1997008298A1 (fr) 1995-08-30 1997-03-06 Genzyme Corporation Purification d'adenovirus et de virus adeno-associe (aav) par voie chromatographique
WO1997009441A2 (fr) 1995-09-08 1997-03-13 Genzyme Corporation Vecteurs aav ameliores pour la therapie genique
WO1997021825A1 (fr) 1995-12-15 1997-06-19 Systemix, Inc. Procede de production de lignees de cellules d'encapsidation retrovirales generant un surnageant retroviral a efficacite de transduction elevee
WO1998009657A2 (fr) 1996-09-06 1998-03-12 Trustees Of The University Of Pennsylvania Methode de therapie genique basee sur des virus adeno-associes de recombinaison
WO1999011764A2 (fr) 1997-09-05 1999-03-11 Targeted Genetics Corporation Procedes de generation de preparations de vecteurs de aav recombinants dont le titre est eleve et qui sont exemptes de virus assistant
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US6258595B1 (en) 1999-03-18 2001-07-10 The Trustees Of The University Of Pennsylvania Compositions and methods for helper-free production of recombinant adeno-associated viruses
WO2001083692A2 (fr) 2000-04-28 2001-11-08 The Trustees Of The University Of Pennsylvania Vecteurs aav recombinants dotes de capsides aav5 et vecteurs aav5 pseudotypes dans des capsides heterologues
US20050053922A1 (en) 2003-06-30 2005-03-10 Schaffer David V. Mutant adeno-associated virus virions and methods of use thereof
US20090202490A1 (en) 2003-06-30 2009-08-13 Schaffer David V Mutant adeno-associated virus virions and methods of use thereof
US9434928B2 (en) 2011-11-23 2016-09-06 Nationwide Children's Hospital, Inc. Recombinant adeno-associated virus delivery of alpha-sarcoglycan polynucleotides
US9614423B2 (en) 2012-04-07 2017-04-04 Traugott Weller Method for producing rotating electrical machines
US9620777B2 (en) 2013-09-30 2017-04-11 Tdk Corporation Positive electrode and lithium ion secondary battery using thereof
US9818600B2 (en) 2014-03-21 2017-11-14 Hitachi Kokusai Electric, Inc. Substrate processing apparatus and method of manufacturing semiconductor device
US9613872B2 (en) 2014-09-29 2017-04-04 Kabushiki Kaisha Toshiba Method of manufacturing semiconductor device
WO2020163300A1 (fr) * 2019-02-04 2020-08-13 Research Institute At Nationwide Children's Hospital Administration de virus adéno-associé de polynucléotide cln3

Non-Patent Citations (51)

* Cited by examiner, † Cited by third party
Title
"Genbank", Database accession no. EU285562
"GenBank", Database accession no. NC_00 1862
BOSCH ET AL., J NEUROSCI, vol. 36, no. 37, 2016, pages 9669 - 9682
BOSCH ET AL., J NEUROSCI., vol. 36, no. 37, 2016, pages 9669 - 9682
CARTER, CURRENT OPINIONS IN BIOTECHNOLOGY, 1992, pages 1533 - 539
CHOUDHURY ET AL., MOL. THER., vol. 24, no. 7, 2016, pages 1247 - 1257
CLARK ET AL., GENE THERAPY, vol. 3, 1996, pages 1124 - 1132
CLARK ET AL., HUM. GENE THER., vol. 10, no. 6, 1999, pages 1031 - 1039
COTMAN ET AL., CLIN LIPIDOL., vol. 7, no. 1, February 2012 (2012-02-01), pages 79 - 91
COTMAN ET AL., HUM MOL GENET, vol. 11, no. 22, 2002, pages 2709 - 2721
COTMAN ET AL., HUM MOL GENET., vol. 11, no. 22, 2002, pages 2709 - 2721
GAO ET AL., J. VIROL., vol. 78, 2004, pages 6381 - 6388
GILES ET AL., MOLECULAR THERAPY, vol. 26, no. 12, 2018, pages 2848 - 2862
HERMONAT ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6466
KITZMII ET AL., HUMAN MOLECULAR GENETICS, vol. 17, no. 2, 2008, pages 303 - 312
KITZMIILLER ET AL., HUM MOL GENET, vol. 17, no. 2, 15 January 2008 (2008-01-15), pages 303 - 12
KITZMIILLER ET AL., HUM MOL GENET., vol. 17, no. 2, 15 January 2008 (2008-01-15), pages 303 - 12
KWON JM ET AL., NEUROLOGY, vol. 77, no. 20, 2011, pages 1801 - 1807
LAUGHLIN ET AL., GENE, vol. 23, 1983, pages 65 - 73
LEBKOWSKI ET AL., MOL. CELL. BIOL., vol. 7, 1988, pages 349
LERNER ET AL., CELL, vol. 82, no. 6, 22 September 1995 (1995-09-22), pages 949 - 57
MARSHALL ET AL., NEUROLOGY, vol. 65, no. 2, 2005, pages 275 - 279
MARSIC ET AL., MOLECULAR THERAPY, vol. 22, no. 11, 2014, pages 1900 - 1909
MCCARTY ET AL., GENE THERAPY, vol. 8, 2001, pages 1248 - 1254
MCLAUGHLIN ET AL., J. VIROL., vol. 62, 1988, pages 1963
MOL. THER., vol. 13, no. 1, 2006, pages 67 - 76
MOLE ET AL., BIOCHIM BIOPHYS ACTA - MOL BASIS DIS., vol. 1852, no. 10, 2015, pages 2237 - 2241
MOLE ET AL., EUR J PAEDIATR NEUROL., vol. 5, 2001, pages 7 - 10
MORGAN ET AL., PLOS ONE, vol. 8, no. 11, 2013, pages e78694
MUNROE PB ET AL., AM J HUM GENET., vol. 61, no. 2, 1997, pages 310 - 316
MUZYCZKA, CURR. TOPICS IN MICROBIAL. AND IMMUNOL., vol. 158, 1992, pages 97 - 129
MUZYCZKA, CURRENT TOPICS IN MICROBIOLOGY AND IMMUNOLOGY, vol. 158, 1992, pages 97 - 129
OSORIO ET AL., GENES BRAIN BEHAV, vol. 8, no. 3, April 2009 (2009-04-01), pages 337 - 345
PALMER ET AL., AM J MED GENET, vol. 42, no. 4, 1992, pages 561 - 567
PALMER ET AL., AM J MED GENET., vol. 42, no. 4, 1992, pages 561 - 567
PAUL ET AL., HUMAN GENE THERAPY, vol. 4, 1993, pages 609 - 615
PERRIN ET AL., VACCINE, vol. 13, 1995, pages 1244 - 1250
PONTIKIS ET AL., NEUROBIOL DIS, vol. 20, no. 3, 2005, pages 823 - 836
RATSCHIN ET AL., MOL. CELL. BIOL., vol. 4, 1984, pages 2072
SAMULSKI ET AL., J. VIROL., vol. 63, 1989, pages 3822 - 3828
SAMULSKI ET AL., PROC. NATL. ACAD. S6. USA, vol. 79, 1982, pages 2077 - 2081
SCHENPPCLARK, METHODS MOL. MED., vol. 69, 2002, pages 427 - 443
SCHULZ A ET AL., BIOCHIMIC ET BIOPHYSICA ACTA, vol. 1832, no. 11, 2013, pages 1801 - 1806
SEEHAFER ET AL., J NEUROIMMUNOL., vol. 230, 2011, pages 169 - 172
SEEHAFER ET AL., NEUROBIOL AGING, vol. 27, 2006, pages 576 - 588
SEIGEL ET AL., MOL CELL NEUROSCI, vol. 19, no. 4, April 2002 (2002-04-01), pages 515 - 27
SENAPATHYCARTER, J. BIOL. CHEM., vol. 259, 1984, pages 4661 - 4666
SPONSOR: "Gene Therapy for Children With CLN3 Batten Disease -Full Text View -ClinicalTrials Study Details Tabular View No Results Posted Disclaimer How to Read a Study Record Gene Therapy for Children With CLN3 Batten Disease", 10 December 2018 (2018-12-10), XP055930052, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/show/NCT03770572?cond=cln3+batten&draw=2&rank=1> [retrieved on 20220610] *
SRIVASTAVA ET AL., J. VIROL., vol. 45, 1983, pages 555 - 564
TRATSCHIN ET AL., MOL. CELL. BIOL., vol. 5, 1985, pages 3251
VIROLOGY, vol. 330, no. 2, 2004, pages 375 - 383

Also Published As

Publication number Publication date
TW202246513A (zh) 2022-12-01

Similar Documents

Publication Publication Date Title
US20210069292A1 (en) Recombinant glut1 adeno-associated viral vector constructs and related methods for restoring glut1 expression
AU2018365677A1 (en) Means and method for preparing viral vectors and uses of same
US11903985B2 (en) Gene therapies for lysosomal disorders
US20220127641A1 (en) Adeno-Associated Virus Delivery of CLN3 Polynucleotide
US20230321164A1 (en) Intrathecal Delivery of Recombinant Adeno-Associated Virus Encoding Methyl-CPG Binding Protein 2
JP2022523766A (ja) Grn関連成人発症性神経変性の治療のための組換えアデノ随伴ウイルス
US20220202956A1 (en) Adeno-associated virus delivery of cln6 polynucleotide
US20230310654A1 (en) Gene therapies for lysosomal disorders
WO2022170038A1 (fr) Administration de virus adéno-associé de polynucléotide cln3
TW202045728A (zh) 用於治療克拉培氏病之組成物
RU2796274C2 (ru) Вирусные векторы на основе aav и пути их применения
BR112021015285A2 (pt) Entrega de vírus adenoassociado de polinucleotídeo cln3
WO2024011115A1 (fr) Administration de polynucléotide cln1 par un virus adéno-associé
CA3216711A1 (fr) Virus adeno-associe recombinant codant pour la proteine 2 de liaison a la methyl-cpg pour traiter le syndrome de pitt hopkins par administration intrathecale
JP2024505257A (ja) 神経セロイドリポフスチン症の遺伝子治療
JP2024515623A (ja) 髄腔内送達によってピット・ホプキンス症候群を治療するためのメチル-cpg結合タンパク質2をコードする組換えアデノ随伴ウイルス
BR112021015150A2 (pt) Entrega de vírus adenoassociado de polinucleotídeo de cln6

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22712101

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22712101

Country of ref document: EP

Kind code of ref document: A1