WO2011072091A1 - Méthodes et compositions utilisées pour le traitement de maladies, d'affections ou de lésions du snc - Google Patents

Méthodes et compositions utilisées pour le traitement de maladies, d'affections ou de lésions du snc Download PDF

Info

Publication number
WO2011072091A1
WO2011072091A1 PCT/US2010/059597 US2010059597W WO2011072091A1 WO 2011072091 A1 WO2011072091 A1 WO 2011072091A1 US 2010059597 W US2010059597 W US 2010059597W WO 2011072091 A1 WO2011072091 A1 WO 2011072091A1
Authority
WO
WIPO (PCT)
Prior art keywords
subject
disease
seq
homo
rna
Prior art date
Application number
PCT/US2010/059597
Other languages
English (en)
Inventor
Elena Feinstein
Igor Spivak
Evgenia Alpert
Ron Lahav
Original Assignee
Quark Pharmaceuticals, 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 Quark Pharmaceuticals, Inc. filed Critical Quark Pharmaceuticals, Inc.
Priority to US13/514,880 priority Critical patent/US8778904B2/en
Priority to EP10809113.3A priority patent/EP2510098B1/fr
Publication of WO2011072091A1 publication Critical patent/WO2011072091A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0046Ear
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific

Definitions

  • the present invention relates to a method of treating a subject at risk of or afflicted with a disease, a disorder or an injury of the central nervous system (CNS), the method comprising administering an otic composition comprising a therapeutically effective amount of an oligonucleotide compound to the ear of the subject.
  • CNS diseases, disorders and injury of the CNS affect millions of people worldwide. With an increase in lifespan and changing population demographics, the incidence of CNS diseases is expected to increase significantly in the 21st century.
  • the delivery of therapeutic oligonucleotide compounds to the CNS for effective treatment of CNS diseases, disorders and injury present a major challenge. For therapeutic purpose, it is important to consider not only the net delivery of a nucleic acid compounds to the CNS, but also the ability of the therapeutic oligonucleotide to access the relevant target site within the CNS.
  • oligonucleotide compounds have been systemic administration, injection into cerebrospinal fluid (CSF) pathways and direct injection into the brain.
  • CSF cerebrospinal fluid
  • modulation of the expression of certain genes involved in Alzheimer's diseases (AD), as well as that of genes involved in Huntington's disease has been attained in vivo by intrathecal and intracerebroventricular administration, implantation of catheters and pumps, by chemical or osmotic opening of the blood-brain barrier, by direct injection or perfusion at the site of injury or lesion, or by direct injection or perfusion into the arterial system of the brain (see for example WO 2005/003350, US 20050042646 and GB 2415961).
  • This disclosure is directed to no n- invasive methods of treating a subject afflicted with a disease, disorder or injury of the central nervous system (CNS) comprising administering to the subject's ear canal an otic composition comprising at least one therapeutic oligonucleotide which targets a gene associated with the disease, disorder or injury .
  • oligonucleotides have allegedly been delivered to the CNS tissue by systemic administration, injection into cerebrospinal fluid (CSF) and direct injection into the brain, as well as by intranasal administration.
  • CSF cerebrospinal fluid
  • Provided herein are otic compositions, and methods of use thereof for treating diseases, disorders and injury of the CNS.
  • the method disclosed herein provides an alternative to invasive delivery of therapeutics to the CNS with greater patient comfort and compliance.
  • RNA compound which down regulates expression of a target gene associated with loss of the retinal ganglion cell, thereby inhibiting loss of the retinal ganglion cell or rescuing a retinal ganglion cell in the subject.
  • the subject is suffering from an ocular disease, an ocular disorder or an ocular injury or at risk of developing an ocular disease, an ocular disorder, or an ocular injury.
  • the ocular disease, disorder, or injury comprises neurodegeneration, increased intraocular pressure and or optic nerve injury.
  • the disease, disorder, or injury is selected from a group consisting of glaucoma, diabetic retinopathy (DR), diabetic macular edema (DME), age related macular degeneration (AMD), Leber's hereditary optic neuropathy (LHON), Leber optic atrophy, optic neuritis, retinal artery occlusion, central retinal vein occlusion, brunch retinal vein occlusion, ischemic optic neuropathy, optic nerve injury, retinopathy of prematurity (ROP) or retinitis pigmentosa (RP), retinal ganglion degeneration, macular degeneration, hereditary optic neuropathy, metabolic optic neuropathy, optic neuropathy due to a toxic agent or neuropathy caused by adverse drug reactions or vitamin deficiency.
  • DR diabetic retinopathy
  • DME diabetic macular edema
  • AMD age related macular degeneration
  • LHON Leber's hereditary optic neuropathy
  • LHON Leber's hereditary optic neuropathy
  • the disease is glaucoma or ischemic optic neuropathy.
  • the otic composition is formulated as a cream, a foam, a paste, an ointment, an emulsion, a liquid solution, an ear drop, a gel, spray, a suspension, a microemulsion, microspheres, microcapsules, nanospheres, nanoparticles, lipid vesicles, liposomes, polymeric vesicles, a patch, or an insert.
  • the otic composition is formulated as an ear drop.
  • the ear drop is topically applied to the ear canal.
  • the ear drop is topically applied to the tympanic membrane.
  • the composition is applied transtympanically.
  • the target gene is set forth in any one of SEQ ID NO: 1-293. In some embodiments the target gene is set forth in any one of SEQ ID NO:22-23.
  • a method of rescuing a retinal ganglion cell from apoptosis in a subject comprising applying to ear canal of the subject an otic composition comprising a therapeutically effective amount of at least one double stranded RNA compound targeting a gene in the retina of the subject, thereby rescuing retinal ganglion cell from apoptosis in the subject.
  • a method for promoting survival of a retinal ganglion cell in a subject displaying signs or symptoms of an ocular neuropathy comprising applying to ear of the subject an otic composition comprising a therapeutically effective amount of at least one double stranded RNA compound to a target gene that promotes survival of a retinal ganglion cell, thereby promoting survival of a retinal ganglion cell in the subject.
  • the signs or symptoms are mediated by apoptosis.
  • kits for preventing, treating or alleviating the effects of an ocular disease associated with death of a retinal ganglion cell in a subject comprising applying to the ear of the subject an otic composition comprising a therapeutically effective amount of at least one double stranded RNA compound to a target gene associated with the ocular disease, thereby preventing, treating or alleviating the effects of an ocular disease associated with death of a retinal ganglion cell in the subject.
  • retinal ganglion cell death is mediated by elevated intraocular pressure (IOP) in the eye of a subject or results from an ischemic event.
  • IOP intraocular pressure
  • a method of delaying, preventing or rescuing a retinal cell from death in a subject suffering from elevated IOP comprising applying to the ear of the subject an otic composition comprising a therapeutically effective amount of at least one double stranded RNA compound to a target gene associated with death of the RGC in the retina of the subject, thereby delaying, preventing or rescuing the retinal cell from injury or death and wherein intraocular pressure (IOP) remains substantially elevated.
  • IOP intraocular pressure
  • the subject is afflicted with glaucoma.
  • a method treating a subject suffering from retinal ganglion cell loss or retinal ganglion cell damage comprising administering to the ear of the subject an otic composition comprising a therapeutically effective amount of at least one double stranded RNA compound to a target gene associated with the retinal ganglion cell loss or damage, thereby treating the subject or reducing retinal ganglion cell death in the subject.
  • a method for attenuating retinal ganglion cell loss and providing ocular neuroprotection to a subject in need thereof comprising applying to the ear of the subject an otic composition comprising a therapeutically effective amount of at least one double stranded RNA compound to a target gene associated with retinal ganglion cell loss, thereby attenuating retinal ganglion cell loss and providing ocular neuroprotection to the subject.
  • Also provided is a method for preventing visual field loss associated with loss of retinal ganglion cells in a subject comprising administering to the ear of the subject an otic composition comprising a therapeutically effective amount of at least one double stranded RNA compound to a target gene in the retina of the subject, thereby preventing visual field loss in the subject.
  • a method of delivering a therapeutic oligonucleotide to the CNS of a subject suffering from a disease, a disorder or an injury of the CNS comprising topically administering to the subject's ear canal an otic composition comprising an effective amount of the oligonucleotide which targets a gene associated with the disease, disorder or injury , and a pharmaceutically acceptable excipient or mixtures thereof, thereby reducing expression of the gene and treating the subject.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from a disease, a disorder or an injury of the CNS, which comprises applying to the subject's ear canal an otic composition comprising an effective amount an oligonucleotide directed to the target gene, and a pharmaceutically acceptable excipient or mixtures thereof, thereby attenuating expression of the target gene in the subject.
  • the CNS injury comprises injury to the retina including retinal ganglion cells (RGC) and/or the optic nerve (ON).
  • the injury to the retina or optic nerve comprises ischemia or hypoxia injury.
  • a method of protecting neuronal cells from cell death in a subject having or at risk of having a neurodegenerative or neurological disease comprising applying to the subject's ear canal a composition comprising a therapeutically effective amount of an oligonucleotide that inhibits a gene associated with the neuronal cell death thereby providing neuroprotection to the neuronal cells.
  • the neuronal cells are present in the CNS.
  • the neuronal cells are cells of the spinal cord.
  • the neuronal cells are retinal ganglion cells or optic nerve cells.
  • the neuronal cells are brain cells.
  • a method of protecting neuronal cells from cell death in a subject having or at risk of having an injury to the CNS comprising applying to the subject's ear canal a composition comprising a therapeutically effective amount of an oligonucleotide that inhibits a gene associated with the neuronal cell death; and a pharmaceutically acceptable excipient or mixtures thereof, thereby providing neuroprotection to the neuronal cells.
  • the injury is injury to the optic nerve and/or retinal ganglion cells.
  • a method of providing neuroprotection from disease or injury to the CNS in a subject in need thereof comprises administering to the subject's ear canal an otic composition comprising at least one oligonucleotide directed to a target gene associated with the neural damage; and a pharmaceutically acceptable excipient or mixtures thereof, thereby reducing expression of the target gene in an amount to afford neuroprotection.
  • a method of promoting neurogenesis or neuroregeneration in a subject in need thereof comprising applying to the subject's ear canal a composition comprising a therapeutically effective amount of an oligonucleotide that inhibits a target gene; and a pharmaceutically acceptable excipient or mixtures thereof, thereby promoting neurogenesis or neuroregeneration in the subject.
  • the target gene is RhoA or a gene within the RhoA pathway.
  • the oligonucleotide is administered in combination with a neurotrophic factor including NGF, BDNF or CNTF.
  • the provided is a method of treating a subject afflicted with a disease, disorder or injury to the CNS, which comprises applying to the subject's ear canal an otic composition comprising at least one oligonucleotide directed to a target gene associated with the disease, disorder or injury; and a pharmaceutically acceptable excipient or mixtures thereof, thereby treating the subject.
  • the disease or disorder comprises intraocular pressure. In some embodiments the disease is glaucoma. In some embodiments the injury is ischemic injury to the optic nerve.
  • a method for reducing neurological inflammation in a subject having or at risk of having a disease, disorder or injury of the CNS comprises applying to the subject's ear canal an otic composition comprising at least one oligonucleotide directed to a target gene associated with the disease, disorder or injury; and a pharmaceutically acceptable excipient or mixtures thereof, thereby reducing neurological inflammation in the subject.
  • a method of treating a subject at risk of retinal degeneration which comprises applying to the subject's ear canal an otic composition comprising at least one oligonucleotide directed to a target gene associated with the retinal degeneration; and a pharmaceutically acceptable excipient or mixtures thereof, thereby reducing the risk of retinal degeneration in the subject.
  • the present invention provides a method of treating a subject suffering from retinal degeneration, which comprises applying to the subject's ear canal an otic composition comprising at least one oligonucleotide directed to a target gene associated with the retinal degeneration; and a pharmaceutically acceptable excipient or mixtures thereof, thereby treating the subject.
  • the target gene is Caspase 2 (CASP2).
  • the present invention provides a method of non-invasive delivery of an oligonucleotide to a retinal tissue in a subject suffering from an eye disorder, disease or injury comprising topically applying an otic composition comprising an oligonucleotide compound to the ear canal of the subject.
  • the present invention provides a method of non-invasive delivery of an oligonucleotide to a retinal ganglion cell in a subject suffering from an eye disorder comprising topically applying an otic composition comprising an oligonucleotide compound to the ear canal of the subject.
  • the present invention provides a method of attenuating expression of a target gene associated with loss of a retinal ganglion cell in the retina in a subject suffering from an ocular disease, disorder or injury, which comprises topically (non-invasively) administering to the ear canal of the subject a pharmaceutical composition comprising at least one oligonucleotide directed to the target mRNA product of the target gene, in an amount and over a period of time effective to attenuate expression of the gene in the retina of the subject.
  • the present invention provides a method of treating a subject suffering from retinal ganglion cell loss or retinal ganglion cell damage and providing ocular neuroprotection to a subject suffering from or at risk of developing an eye disease, disorder or injury.
  • the method comprises topically administering to the ear canal of the subject an otic pharmaceutical composition comprising at least one oligonucleotide directed to a target gene in the retina of the subject, in an amount and over a period of time effective to inhibit retinal ganglion cell loss or retinal ganglion cell damage in the subject.
  • the CNS injury results from exposure to a neurotoxin.
  • the invention provides a method of treating neurotoxicity in a subject in need thereof, which comprises administering to the subject's ear canal an otic pharmaceutical composition comprising at least one oligonucleotide directed to a target gene associated with neurotoxicity in the CNS, and a pharmaceutically acceptable excipient or mixtures thereof, thereby reducing expression of a gene associated with the neurotoxicity in the CNS of the subject in an amount and over a period of time effective to treat the subject.
  • the otic pharmaceutical compositions further comprises a permeability enhancer, also known as penetration enhancer.
  • the present invention provides a method of treating a subject suffering from or at risk of a disease, a disorder or an injury of the CNS which comprises topically administering to the canal of the subject's ear an otic pharmaceutical composition comprising: (a) a therapeutically effective amount of at least one oligonucleotide compound which inhibits the expression of a human target gene associated with a disease, a disorder or an injury of the CNS; (b) a permeability enhancer and (c) a pharmaceutically acceptable excipient or carrier, or mixtures thereof, thereby treating the subject.
  • the penetration enhancer is selected from any compound or any combination of two ore more compounds that enhance the penetration of a therapeutic oligonucleotide through the skin and/or the tympanic membrane in the ear of a subject suffering from or at risk of a disease, a disorder or an injury of the CNS.
  • the permeability enhancer is a polyol.
  • the oligonucleotide is in admixture with a polyol.
  • the polyol is selected from the group consisting of glycerol, propylene glycol, polyethylene glycol, sorbitol, xylitol, maltitol and combinations thereof.
  • the polyol is glycerol.
  • glycerol is present at a final concentration of about 0.1% to about 35%; about 1% to about 30%; about 5% to about 25%, preferably about 10% to about 20% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 2%, 2.5%, 5%, 10%, 12.5%, 15%, 17.5%, 20%, 22.5%, 25%, 27.5% or about 30% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 2% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 10% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 20% by volume of the otic pharmaceutical composition.
  • the pharmaceutical composition is brought to the subject's body temperature, which is about 30°C to about 38°C, prior to application to the ear.
  • the pharmaceutical composition is applied to the ear canal when the subject's head is tilted to one side and the treated ear is facing upward.
  • the pharmaceutical composition is applied to the ear using a receptacle for eardrops, for example using a dropper of for example, 10-100 microliter per drop, or a wick.
  • the at least one oligonucleotide compound is selected from chemically modified double stranded RNA, unmodified double stranded RNA, antisense, ribozyme, miRNA and shRNA compound. In preferred embodiments the at least one oligonucleotide is a chemically modified double stranded RNA compound.
  • the target mRNA is a product of a gene selected from the group consisting of APP, MAPT, SOD1, BACE1, CASP3, TGM2, TARDBP, ADRB1, CAMK2A, CBLN1, CDK5R1, GABRA1, MAPK10, NOS1, NPTX2, NRGN, NTS, PDCD2, PDE4D, PENK, SYT1, TTR, FUS, LRDD, CYBA, ATF3, CASP2, HRK, C1QBP, BNIP3, MAPK8, MAPK14, Racl, GSK3B, P2RX7, TRPM2, PARG, CD38, STEAP4, BMP2, GJA1, TYROBP, CTGF, ANXA2, DUOX1, RTP801, RTP801L, NOX4, NOX1, NOX2 (gp91pho, CYBB), NOX5, DUOX2, NOXOl, NOX02 (p47phox, NCF1), NOXA1, NOX
  • Attenuating expression of at least one target mRNA confers upon the cells and/or tissues of the CNS neuroprotective properties.
  • APP MAPT, SOD1, BACE1, CASP3, TGM2, TARDBP, ADRB1, CAMK2A, CBLN1, CDK5R1, GABRA1, MAPK10, NOS1, NPTX2, NRGN, NTS, PDCD2, PDE4D, PENK, SYT1, TTR, FUS, LRDD, CYBA, ATF3, CASP2, HRK, C1QBP, BNIP3, MAPK8, MAPK14, Racl, GSK3B, P2RX7, TRPM2, PARG, CD38, STEAP4, BMP2, GJA1, TYROBP, CTGF, ANXA2, DUOX1, RTP801, RTP801L, NOX4, NOX1, NOX2 (gp91pho, CYBB), NOX5, DUOX2, NOXOl, NOX02 (p47phox, NCF1), NOXA1, NOXA2 (p67phox, NCF2), p53 (TP53), HTRA2, KEAP1,
  • inhibiting expression of at least one target gene confers upon the cells and/or tissues of the CNS neuroprotective properties.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the at least one target gene is selected from a gene transcribed into an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the otic oligonucleotide composition is administered to the external ear, including the ear canal of a subject by any suitable mode of administration.
  • suitable modes of administration of the otic oligonucleotide compositions of the invention include invasive and non-invasive modes of administration, such as without being limited to, instillation (of ear drops), injection, deposition, or spraying into the ear.
  • the compositions of the present invention are administered topically into the ear canal as eardrops or injected through a cannula into the ear canal or injected through the tympanic membrane (transtympanic injection).
  • the method employs applying the pharmaceutical composition to the subject's ear canal when the subject's head is tilted to one side and the treated ear is facing upward.
  • the pharmaceutical composition is applied to the ear using a receptacle for eardrops, for example using a dropper of for example, 10-100 microliter per drop, or a wick.
  • the compositions of the present invention are brought to the subject's body temperature, which is about 30°C to about 38°C, prior to application to the ear.
  • the at least one double stranded RNA compound is delivered to the ear of a subject in a pharmaceutical composition formulated as an eardrop.
  • a non-invasive method of attenuating expression of a target mRNA in a subject suffering from a disease, a disorder or an injury of the CNS which comprises topically administering into the ear canal of the subject a pharmaceutical composition formulated as an ear drop comprising at least one oligonucleotide directed to the target mRNA, in an amount and over a period of time effective to attenuate expression of the target mRNA in the CNS of the subject.
  • the present invention provides a method of treating a disease, a disorder or an injury of the CNS in a subject in need thereof, which comprises topically administering to the ear canal of the subject a pharmaceutical composition formulated as an ear drop, comprising at least one oligonucleotide directed to a target gene associated with the disease, the disorder or the injury of the CNS, in an amount and over a period of time effective to treat the subject.
  • the disease, disorder or injury of the central nervous system is selected from eye disorder, neurodegenerative disease, spinal cord disease, traumatic and nontraumatic spinal cord injury, traumatic brain injury, cancer in the central nervous system (CNS), neurological disorder, mood disorders and other diseases associated with inflammation and / or neurotoxicity and / or oxidative stress in the CNS.
  • the eye disorder, disease or injury is selected from glaucoma, diabetic retinopathy (DR), diabetic macular edema (DME), age related macular degeneration (AMD) Leber's hereditary optic neuropathy (LHON) or Leber optic atrophy .
  • the disorder is a primary glaucoma, selected from primary open angle glaucoma, normal- tension glaucoma or angle-closure glaucoma.
  • the disorder is a secondary glaucoma selected from pseudoexfoliation glaucoma, pigmentary glaucoma, neovascular glaucoma, steroid-induced glaucoma or treatment refractory glaucoma.
  • the ocular disorder, disease or injury is optic neuritis, retinal artery occlusion, central retinal vein occlusion, brunch retinal vein occlusion (BRVO).
  • the eye disorder, disease or injury is retinitis pigmentosa (RP), ischemic optic neuropathy or optic nerve injury.
  • RP retinitis pigmentosa
  • the optic neuropathy is selected from non-arteritic anterior ischemic optic neuropathy (NAION), optic neuritis, neuromyelitis optica, dominant optic atrophy, Leber's hereditary optic neuropathy.
  • ocular disorder, disease or injury is retinopathy of prematurity (ROP) retinal ganglion degeneration, macular degeneration, hereditary optic neuropathy, metabolic optic neuropathy, optic neuropathy due to a toxic agent or neuropathy caused by adverse drug reactions or vitamin deficiency.
  • ROP retinopathy of prematurity
  • the disorder is vision loss associated with a tumor.
  • the neurodegenerative disorder is selected from neurodegenerative conditions causing problems with movements, such as impairment of motor, sensory or autonomic function; and conditions affecting memory and related to cognitive impairment or dementia.
  • the neurodegenerative disorder is selected from, without being limited to, Parkinson's disease, Amyotrophic Lateral Sclerosis (ALS, also referred to as Lou Gehrig's Disease), Prion disease dementia, amnestic mild cognitive impairment, Alzheimer's disease, Lewy body dementia, Pick's disease, Ataxia-telangiectasia (AT), Frontotemporal dementia (FTD), Frontotemporal lobar degeneration (FTLD), Huntington's disease and any other disease- induced dementia (including, HIV-associated dementia and post- stroke dementia, for example).
  • Parkinson's disease Amyotrophic Lateral Sclerosis
  • ALS also referred to as Lou Gehrig's Disease
  • Prion disease dementia amnestic mild cognitive impairment
  • Alzheimer's disease Lewy body dementia
  • Pick's disease Ataxia-telangiectasia
  • FDD Frontotempo
  • the disorder is a neurological disorder selected from, without being limited to, no n- traumatic neurological disease that affects the spinal cord, stroke, epilepsy, Parkinsonism, Gluten Ataxia, cerebral ischemia and cerebrovascular accident.
  • the disease or disorder is a neoplasm in the CNS selected from any intracranial tumor created by abnormal and uncontrolled cell division either in the brain itself or spread from cancers primarily located in other organs (i.e. metastatic tumors).
  • the neoplasm in the CNS is created by abnormal proliferation of or in the, inter alia, neurons (e.g. Motor neuron, Purkinje neuron, GABAergic neuron, Multipolar neuron, Cerebellar neuron, Afferent neuron, Sensory neuron), glial cells (e.g.
  • astrocytes astrocytes, microglia, oligodendrocytes), ependymal cells, lymphatic tissue, blood vessels, cranial nerves, myelin- producing Schwann cells, meninges, skull, Striatum, Nucleus of stria terminalis, hypothalamus, pituitary gland and pineal gland.
  • the neoplasm in the CNS is an intracranial glioma selected from, without being limited to, ependymoma, glioma, astrocytoma, oligodendroglioma and oligoastrocytoma.
  • the intracranial glioma is selected from Pilocytic astrocytoma of cerebellum and Oligodendroglioma of brain.
  • the neoplasm is a neural crest tumor such as e.g. cranial primitive neuroectodermal tumors (PNET).
  • PNET cranial primitive neuroectodermal tumors
  • the neoplasm is selected from, without being limited to, Medulloblastoma of cerebellum, Neuroblastoma of brain, Glioblastoma multiforme of brain and Neurofibromatosis.
  • the disease or disorder of the CNS is selected from, without being limited to, Supranuclear paralysis, Lymphocytic choriomeningitis, Niemann Pick disease (e.g. Niemann Pick disease Type C) and AF type amyloidosis (Familial neuropathic amyloidosis).
  • the injury of the CNS is selected from, without being limited to, traumatic and non-traumatic spinal cord injury, and brain injury (e.g. Traumatic Brain Injury (TBI)), that is caused by fracture or penetration of the skull (e.g. a vehicle accident, fall, gunshot wound), a disease process (e.g. neurotoxins, infections, tumors, metabolic abnormalities, etc.) or a closed head injury such as in the case of rapid acceleration or deceleration of the head (e.g. Shaken Baby Syndrome, blast), blunt trauma, concussions, and concussion syndrome.
  • TBI Traumatic Brain Injury
  • the disease or disorder of the CNS is selected from mood disorders (e.g. major depressive disorder and bipolar disorder) and Post-traumatic stress disorder (PTSD).
  • mood disorders e.g. major depressive disorder and bipolar disorder
  • PTSD Post-traumatic stress disorder
  • the disease, disorder or injury of the central nervous system is selected from a disease associated with inflammation and / or neurotoxicity and / or oxidative stress in the CNS, such as, without being limited to, demyelinating disease (e.g. Multiple Sclerosis, Acute Inflammatory Demyelinating Polyradiculoneuropathy (AIDP), Chronic Inflammatory demyelinating polyradiculoneuropathy (CIDP), Guillain-Barre syndrome (GBS)).
  • demyelinating disease e.g. Multiple Sclerosis, Acute Inflammatory Demyelinating Polyradiculoneuropathy (AIDP), Chronic Inflammatory demyelinating polyradiculoneuropathy (CIDP), Guillain-Barre syndrome (GBS)
  • the present invention provides an otic pharmaceutical composition
  • an otic pharmaceutical composition comprising: (a) a therapeutically effective amount of at least one oligonucleotide compound which inhibits the expression of a human target gene associated with a disease, a disorder or an injury of the CNS, wherein the target gene is selected from one or more of: APP, MAPT, SOD1, BACE1, CASP3, TGM2, TARDBP, ADRB1, CAMK2A, CBLN1, CDK5R1, GABRA1, MAPK10, NOS1, NPTX2, NRGN, NTS, PDCD2, PDE4D, PENK, SYT1, TTR, FUS, LRDD, CYBA, ATF3, CASP2, HRK, C1QBP, BNIP3, MAPK8, MAPK14, Racl, GSK3B, P2RX7, TRPM2, PARG, CD38, STEAP4, BMP2, GJA1, TYROBP, CTGF, ANXA2, DUOX1, RTP801,
  • the at least one oligonucleotide compound is a chemically double stranded RNA compound. In some preferred embodiments the at least one oligonucleotide compound is a chemically modified siRNA.
  • Otic pharmaceutical composition of the invention is useful with any oligonucleotide pair (sense and antisense strands) to a mammalian gene or non-mammalian gene.
  • the mammalian gene is a human gene.
  • the non- mammalian gene is involved in a mammalian disease, preferably human disease.
  • the human gene is selected from the group consisting of genes having mRNA set forth inany one of SEQ ID NOS: 1-293. Examples of oligonucleotide sequence pairs are provided in PCT Patent Publication Nos.
  • the penetration enhancer is selected from any compound or any combination of two ore more compounds that enhance the penetration of a therapeutic oligonucleotide through the skin and/or the tympanic membrane in the ear of a subject suffering from or at risk of a disease, a disorder or an injury of the CNS.
  • the permeability enhancer is a polyol.
  • the oligonucleotide is in admixture with a polyol.
  • the polyol is selected from the group consisting of glycerol, propylene glycol, polyethylene glycol, sorbitol, xylitol, maltitol, and combinations thereof.
  • the polyol is glycerol.
  • glycerol is present at a final concentration of about 0.1% to about 35%; about 1% to about 30%; about 5% to about 25%, preferably about 10% to about 20% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 2%, 2.5%, 5%, 10%, 12.5%, 15%, 17.5%, 20%, 22.5%, 25%, 27.5% or about 30% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 2% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 10% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 20% by volume of the otic pharmaceutical composition.
  • the otic pharmaceutical composition is brought to the subject's body temperature, which is about 30°C -38°C, prior to application to the ear.
  • FIG. 1 CASP2_4_S510 siRNA (siCASP2) hybridization signals in brain section, 6h post ErD application of siRNA (#53).
  • Example 4 CASP2_4_S510 siRNA (siCASP2) hybridization signals in brain section, 6h post ErD application of siRNA (#53).
  • FIG. 2 Lateral ventricle and adjacent tissue (#54. Original magnification xlO. Exposure 6 days). When siRNA appeared in ventricles (probably through cerebrospinal fluid CSF), it was also absorbed into the adjacent brain tissue (hippocampus, striatum, thalamus). (Example 4)
  • FIG. 3 CASP2_4_S510 siRNA signals in spinal trigeminal tract, principal sensory 5 nucleus, vestibule-cochlear nerve 8n, (#54. Original magnification A x2.5, B & C xlO, exposure Id). (Example 4)
  • FIG. 4 CASP2_4_S510 siRNA signal in facial nerve - 7n (#54. Original magnification xlO. Exposure Id).
  • Figure 5 CASP2 gene knockdown (bar designated as "siGeneX”) in the rat retina at 24 hours after administration of otic composition administered via eardrops (Experimental Group II).
  • CNL_1 ((Experimental Group IV) is identified as "siContr”
  • 10% glycerol group ((Experimental Group VI) is identified as "Vehicle”
  • intact group Experimental Group VII) is identified "Intact”.
  • Example 7 Example 7
  • the present invention provides topical oligonucleotide compositions, in particular, otic oligonucleotide compositions, and methods of use thereof for treating various diseases, disorders and injury of the Central Nervous System (CNS).
  • the present invention is based in part on the surprising finding that auricular/otic administration of double stranded RNA compositions targets certain tissues and cell types of Central Nervous System (CNS).
  • the finding is surprising in view of the known difficulties associated with delivery of therapeutic oligonucleotide compounds to the CNS for effective treatment of CNS diseases, disorders and injury.
  • the present invention now discloses non-invasive methods of treating CNS diseases, disorders and injury.
  • the present invention relates in general to otic pharmaceutical compositions that comprise a therapeutically effective amount of at least one oligonucleotide compound, which inhibits the expression of a target gene associated with a disease, a disorder or an injury of the CNS and to the use of these novel compositions in the treatment of a subject suffering from medical conditions associated with expression of those genes in CNS tissues and cells.
  • the oligonucleotide compound is a double stranded RNA compound, such as small interfering RNA (siRNA).
  • siRNA small interfering RNA
  • Otic pharmaceutical composition of the invention is useful with any oligonucleotide pair (sense and antisense strands) to a mammalian gene or non-mammalian gene.
  • the mammalian gene is a human gene.
  • the non-mammalian gene is involved in a mammalian disease, preferably human disease.
  • the double stranded RNA compounds possess structures and modifications which increase activity, increase stability, minimize toxicity, reduce off target effects and/or reduce immune response when compared to an unmodified double stranded RNA compound; the modifications are beneficially applied to double stranded oligonucleotide sequences useful in preventing or attenuating target gene expression, in particular the target genes discussed herein.
  • the present invention provides a method of treating a disease, a disorder or an injury of the CNS in a subject in need thereof, which comprises administering to the ear of the subject an otic pharmaceutical composition comprising at least one oligonucleotide directed to a target gene associated with the disease, the disorder or the injury of the CNS, in an amount and over a period of time effective to treat the subject.
  • the subject is a human subject.
  • Methods and otic pharmaceutical compositions which inhibit target genes associated with a disease, a disorder or an injury of the CNS, are discussed herein at length.
  • Diseases and conditions to be treated include but are not limited to a neurodegenerative disease, a neurological disorder, a malignancy or a tumor, an affective disorder, or nerve damage resulting from a cerebrovascular disorder, injury or infection of the CNS.
  • the disease or condition of the CNS is a CNS disease associated with inflammation, a CNS disease associated with neurotoxicity, a disease associated with an oxidative stress in the CNS.
  • the target gene associated with a disease, a disorder or an injury of the CNS is selected from one or more of target genes are presented in Tables A, hereinbelow, i.e. APP, MAPT, SOD1, BACE1, CASP3, TGM2, TARDBP, ADRB1, CAMK2A, CBLN1, CDK5R1, GABRA1, MAPK10, NOS1, NPTX2, NRGN, NTS, PDCD2, PDE4D, PENK, SYT1, TTR, FUS, LRDD, CYBA, ATF3, CASP2, HRK, C1QBP, BNIP3, MAPK8, MAPK14, Racl, GSK3B, P2RX7, TRPM2, PARG, CD38, STEAP4, BMP2, GJA1, TYROBP, CTGF, ANXA2, DUOX1, RTP801, RTP801L, NOX4, NOX1, NOX2 (gp91pho, CYBB), NOX5, DUOX2,
  • Table A mRNA of target genes for certain embodiments of the present invention
  • MTT protein tau
  • gil294862257lreflNM_001123066.31 Homo sapiens microtubule-associated protein tau (MAPT), transcript variant 6, mRNA
  • BACEl beta- site APP-cleaving enzyme 1
  • transcript variant a mRNA
  • caspase 1 apopto sis-related cysteine peptidase (interleukin 1, beta, convertase) (CASP1), transcript variant alpha, mRNA
  • caspase 1 apopto sis-related cysteine peptidase (interleukin 1, beta, convertase) (CASP1), transcript variant beta, mRNA
  • caspase 1 apopto sis-related cysteine peptidase (interleukin 1, beta, convertase) (CASP1), transcript variant gamma, mRNA
  • caspase 2 apopto sis-related cysteine peptidase (neural precursor cell expressed, developmentally down-regulated 2) (CASP2), transcript variant 1, mRNA
  • CASP3 ⁇ SEQ_ID_NO:24;RNA;Homo_Sapiens> GII73622121lreflNM_004346.3l Homo sapiens caspase 3, apopto sis-related cysteine peptidase (CASP3), transcript variant alpha, mRNA
  • TGM2 ⁇ SEQ_ID_NO:28;RNA;Homo_Sapiens> GII39777596lreflNM_004613.2l
  • transglutaminase 2 C polypeptide, protein-glutamine- gamma-glutamyltransferase) (TGM2), transcript variant 1, mRNA
  • Homo sapiens transglutaminase 2 C polypeptide, protein-glutamine- gamma-glutamyltransferase) (UGM2), transcript variant 2, mRNA
  • TARDB ⁇ SEQ_ID_NO:30;RNA;Homo_Sapiens> GII42741653lreflNM_007375.3l P Homo sapiens UAR DNA binding protein (TARDBP), mRNA
  • CAMK2 ⁇ SEQ_ID_NO:32;RNA;Homo_Sapiens> GII212549564lreflNM_015981.3l
  • transcript variant 1 mRNA
  • CBLN1 Homo sapiens cerebellin 1 precursor
  • CDK5R ⁇ SEQ_ID_NO:35;RNA;Homo_Sapiens> GII34304373lreflNM_003885.2l 1 Homo sapiens cyclin- dependent kinase 5, regulatory subunit 1 (p35)
  • GAB A Homo sapiens gamma-aminobutyric acid
  • GABA gamma-aminobutyric acid
  • GABA gamma-aminobutyric acid
  • GABA gamma-aminobutyric acid
  • GABA gamma-aminobutyric acid
  • GABA gamma-aminobutyric acid
  • GABA gamma-aminobutyric acid
  • MAPK1 ⁇ SEQ_ID_NO:43;RNA;Homo_Sapiens> GII257467587lreflNM_002753.3l
  • mitogen-activated protein kinase 10 MAPK10
  • transcript variant 1 mRNA
  • MAPK10 mitogen-activated protein kinase 10
  • MAPK10 mitogen-activated protein kinase 10
  • transcript variant 3 mRNA
  • nitric oxide synthase 1 neurovascular
  • NPTX2 ⁇ SEQ_ID_NO:48;RNA;Homo_Sapiens> GII223671935lreflNM_002523.2l
  • NPTX2 neuronal pentraxin II
  • NRGN Homo sapiens neurogranin (protein kinase C substRte, RC3) (NRGN), transcript variant 1, mRNA
  • GII187131238lreflNM_001126181.1l Homo sapiens neurogranin (protein kinase C substRte, RC3) (NRGN), transcript variant 2, mRNA
  • NTS neurotensin
  • PDCD2 ⁇ SEQ_ID_NO:52;RNA;Homo_Sapiens> GII21735591lreflNM_002598.2l
  • PDCD2 programmed cell death 2
  • transcript variant 1 mRNA
  • PDE4A ⁇ SEQ_ID_NO:54;RNA;Homo_Sapiens> GII162329607lreflNM_001111307.1l Homo sapiens phosphodiesterase 4A, c AMP-specific (phosphodiesterase E2 dunce homolog, Drosophila) (PDE4A), transcript variant 1, mRNA
  • GII162329609lreflNM_001111308.1l Homo sapiens phosphodiesterase 4A, cAMP-specific (phosphodiesterase E2 dunce homolog, Drosophila) (PDE4A), transcript variant 2, mRNA
  • GII259906419lreflNM_001165899.1l Homo sapiens phosphodiesterase 4D, cAMP-specific (phosphodiesterase E3 dunce homolog, Drosophila) (PDE4D), transcript variant 3, mRNA
  • GII208879445lreflNM_001135690.1l Homo sapiens proenkephalin (PENK), transcript variant 1, mRNA
  • Synaptotagmin I STYT1
  • transcript variant 1 mRNA
  • GII209447069lreflNM_001135805.1l Homo sapiens synaptotagmin I (SYT1), transcript variant 2, mRNA
  • GII209447072lreflNM_001135806.1l Homo sapiens synaptotagmin I (SYT1), transcript variant 3, mRNA
  • TTR ⁇ SEQ_ID_NO:66;RNA;Homo_Sapiens> GII221136767lreflNM_000371.3l
  • TTR transthyretin
  • FUS FUS ⁇ SEQ_ID_NO:67;RNA;Homo_Sapiens> GII270265814lreflNM_004960.3l Homo sapiens fused in sarcoma (FUS), transcript variant 1, mRNA
  • GII283135200lreflNM_001170634.1l Homo sapiens fused in sarcoma (FUS), transcript variant 3, mRNA
  • GII283135172lreflNM_001170937.1l Homo sapiens fused in sarcoma (FUS), transcript variant 4, mRNA
  • LRDD leucine-rich repeats and death domain containing
  • ATF3 Homo sapiens activating transcription factor 3 (ATF3), transcript variant 1, mRNA
  • GII95102484lreflNM_001030287.21 Homo sapiens activating transcription factor 3 (ATF3), transcript variant 3, mRNA
  • GII95102482lreflNM_001040619.1l Homo sapiens activating transcription factor 3 (ATF3), transcript variant 4, mRNA
  • C1QBP complement component 1, q subcomponent binding protein
  • Homo sapiens BCL2/adenovirus EIB 19kDa interacting protein 3 (BNIP3), nuclear gene encoding mitochondrial protein, mRNA
  • MAPK8 ⁇ SEQ_ID_NO:81;RNA;Homo_Sapiens> GII20986522lreflNM_139049.1l
  • mitogen-activated protein kinase 8 MAPK8
  • transcript variant 1 mRNA ⁇ SEQ_ID_NO:82;RNA;Homo_Sapiens> GII20986493lreflNM_002750.2l
  • mitogen-activated protein kinase 8 MAPK8
  • transcript variant 2 mRNA
  • mitogen-activated protein kinase 8 MAPK8
  • transcript variant 3 mRNA
  • mitogen-activated protein kinase 8 MAPK8
  • transcript variant 4 mRNA
  • MAPK1 ⁇ SEQ_ID_NO:85;RNA;Homo_Sapiens> GII194578902lreflNM_001315.2l
  • mitogen-activated protein kinase 14 (MAPK14), transcript variant 1, mRNA
  • ras-related C3 botulinum toxin substrate 1 rho family, small GUP binding protein Racl) (RAC1), transcript variant Racl, mRNA
  • GSK3B ⁇ SEQ_ID_NO:91;RNA;Homo_Sapiens> GII225903415lreflNM_002093.3l
  • GSK3B glycogen synthase kinase 3 beta
  • transcript variant 1 mRNA
  • GII225903436lreflNM_001146156.1l Homo sapiens glycogen synthase kinase 3 beta (GSK3B), transcript variant 2, mRNA
  • TRPM2 ⁇ SEQ_ID_NO:94;RNA;Homo_Sapiens> GII67906812lreflNM_003307.3l
  • transient receptor potential cation channel subfamily M, member 2 (TRPM2), transcript variant L, mRNA
  • PARG Homo sapiens poly (ADP-ribose) glycohydrolase
  • CD38 ⁇ SEQ_ID_NO:96;RNA;Homo_Sapiens> GII38454325lreflNM_001775.2l Homo sapiens CD38 molecule (CD38), mRNA
  • STEAP4 Homo sapiens STEAP family member 4 (STEAP4), mRNA
  • BMP2 bone morphogenetic protein 2
  • GJA1 ⁇ SEQ_ID_NO:99;RNA;Homo_Sapiens> GII122939163lreflNM_000165.3l
  • GJA1 Homo sapiens gap junction protein, alpha 1, 43kDa (GJA1), mRNA
  • TYROB ⁇ SEQ_ID_NO: 100;RNA;Homo_Sapiens> GII291045269lreflNM_003332.3l
  • TYROBP Homo sapiens TYRO protein tyrosine kinase binding protein
  • TYROBP TYRO protein tyrosine kinase binding protein
  • GII291045271lreflNM_001173514.1l Homo sapiens UYRO protein tyrosine kinase binding protein (TYROBP), transcript variant 3, mRNA
  • GII291045273lreflNM_001173515.1l Homo sapiens UYRO protein tyrosine kinase binding protein (TYROBP), transcript variant 4, mRNA
  • CTGF connective tissue growth factor
  • GII216547999lreflNM_001002858.2l Homo sapiens annexin A2 (ANXA2), transcript variant 1, mRNA
  • Homo sapiens ras homolog gene family, member A (RHOA), mRNA Homo sapiens ras homolog gene family, member A (RHOA), mRNA.
  • DUOX1 dual oxidase 1
  • transcript variant 1 mRNA
  • DDIT4 ⁇ SEQ_ID_NO: 114;RNA;Homo_Sapiens> GII56676369lreflNM_019058.2l (RTP801 Homo sapiens DNA-damage-inducible transcript 4 (DDIT4), mRNA
  • DDIT4L ⁇ SEQ_ID_NO: 115;RNA;Homo_Sapiens> GII34222182lreflNM_145244.2l (RTP801 Homo sapiens DNA-damage-inducible transcript 4- like (DDIT4L), mRNA L)
  • NADPH oxidase 4 NOX4
  • transcript variant 1 mRNA
  • GII219842345lreflNM_001143836.1l Homo sapiens NADPH oxidase 4 (NOX4), transcript variant 2, mRNA
  • GII219842347lreflNM_001143837.1l Homo sapiens NADPH oxidase 4 (NOX4), transcript variant 3, mRNA
  • NADPH oxidase 1 NOX1
  • transcript variant NOH-1L transcript variant NOH-1L
  • NADPH oxidase organizer 1 NOXOl
  • transcript variant a mRNA
  • Homo sapiens neutrophil cytosolic factor 1, (chronic granulomatous disease, autosomal 1) (NCF1), mRNA (also p47phox, NOX02) NOXA1 ⁇ SEQ_ID_NO: 127;RNA;Homo_Sapiens> GII41393186lreflNM_006647.1l
  • NOXA1 neutrophil cytosolic factor 1
  • NCF2 ⁇ SEQ_ID_NO: 128;RNA;Homo_Sapiens> GII189083740lreflNM_000433.3l (p67pho Homo sapiens neutrophil cytosolic factor 2 (NCF2), transcript variant 1, x, mRNA
  • TP53 Homo sapiens tumor protein p53 (TP53), transcript variant 1, mRNA
  • GII187830776lreflNM_001126112.1l Homo sapiens tumor protein p53 (TP53), transcript variant 2, mRNA
  • GII187830854lreflNM_001126114.1l Homo sapiens tumor protein p53 (TP53), transcript variant 3, mRNA
  • GII187830822lreflNM_001126113.1l Homo sapiens tumor protein p53 (TP53), transcript variant 4, mRNA
  • GII187830893lreflNM_001126115.1l Homo sapiens tumor protein p53 (TP53), transcript variant 5, mRNA
  • GII187830900lreflNM_001126116.1l Homo sapiens tumor protein p53 (TP53), transcript variant 6, mRNA
  • GII187830908lreflNM_001126117.1l Homo sapiens tumor protein p53 (TP53), transcript variant 7, mRNA
  • HTRA2 ⁇ SEQ_ID_NO: 137;RNA;Homo_Sapiens> GII73747817lreflNM_013247.4l
  • HtrA serine peptidase 2 HTRA2
  • nuclear gene encoding mitochondrial protein HtrA serine peptidase 2 (HTRA2)
  • nuclear gene encoding mitochondrial protein HtrA serine peptidase 2 (HTRA2)
  • transcript variant 1 mRNA
  • KEAPl Homo sapiens kelch-like ECH-associated protein 1 (KEAPl), transcript variant 1, mRNA
  • Homo sapiens SHC (Src homology 2 domain containing) transforming protein 1 (SHC1), transcript variant 1, mRNA ⁇ SEQ_ID_NO: 142;RNA;Homo_Sapiens> GII194239660lreflNM_003029.4l
  • Homo sapiens SHC (Src homology 2 domain containing) transforming protein 1 (SHC1), transcript variant 2, mRNA
  • GII194239663lreflNM_001130040.1l Homo sapiens SHC (Src homology 2 domain containing) transforming protein 1 (SHC1), transcript variant 3, mRNA
  • LGALS3 ⁇ SEQ_ID_NO: 146;RNA;Homo_Sapiens> GII294345473lreflNM_002306.3l
  • LGALS3 Homo sapiens lectin, galactoside-binding, soluble, 3 (LGALS3), transcript variant 1, mRNA
  • GII294345474lreflNM_001177388.1l Homo sapiens lectin, galactoside- binding, soluble, 3 (LGALS3), transcript variant 3, mRNA
  • CSD Homo sapiens cathepsin D
  • CAPNS1 Homo sapiens calpain, small subunit 1 (CAPNS1), transcript variant 1, mRNA
  • GII51599150lreflNM_001003962.1l Homo sapiens calpain, small subunit 1 (CAPNS1), transcript variant 2, mRNA
  • Fas TNF receptor superfamily, member 6
  • FAS Fas (TNF receptor superfamily, member 6)
  • transcript variant 1 mRNA
  • RNA RNA;Homo_Sapiens> GII253970389lreflNR_028033.1l Homo sapiens Fas (TNF receptor superfamily, member 6) (FAS), transcript variant 5, non-coding RNA
  • RNA RNA;Homo_Sapiens> GII253970390lreflNR_028034.1l Homo sapiens Fas (TNF receptor superfamily, member 6) (FAS), transcript variant 6, non-coding RNA
  • RNA RNA;Homo_Sapiens> GII253970391lreflNR_028035.1l Homo sapiens Fas (TNF receptor superfamily, member 6) (FAS), transcript variant 7, non-coding RNA
  • Fas ligand TNF superfamily, member 6
  • FASLG Fas ligand
  • calpain 1, (mu/I) large subunit (CAPN1) Homo sapiens calpain 1, (mu/I) large subunit (CAPN1), mRNA
  • TNFRSF6 Homo sapiens Fas (TNFRSF6)-associated via death domain (FADD), mRNA
  • NGFR nerve growth factor receptor
  • mRNA also p75NTR
  • RTN4 ⁇ SEQ_ID_NO: 169;RNA;Homo_Sapiens> GII47519458lreflNM_020532.4l (NogoA) Homo sapiens reticulon 4 (RTN4), transcript variant 1, mRNA
  • RTN4R ⁇ SEQ_ID_NO: 174;RNA;Homo_Sapiens> GII47519383lreflNM_023004.5l (NGR) Homo sapiens reticulon 4 receptor (RTN4R), mRNA
  • MAG myelin associated glycoprotein
  • MAG myelin associated glycoprotein
  • oligodendrocyte myelin glycoprotein OMG
  • PTPRZ1 phospha Homo sapiens protein tyrosine phosphatase, receptor- type, Z polypeptide 1 can
  • TNC ⁇ SEQ_ID_NO: 182;RNA;Homo_Sapiens> GII153946394lreflNM_002160.2l tenascin Homo sapiens tenascin C (TNC), mRNA
  • NBP1 neuropilin 1
  • transcript variant 1 mRNA
  • GII182509170lreflNM_001024628.2l Homo sapiens neuropilin 1 (NRP1), transcript variant 2, mRNA
  • GII182509171lreflNM_001024629.2l Homo sapiens neuropilin 1 (NRP1), transcript variant 3, mRNA
  • NBP2 neuropilin 2
  • transcript variant 1 mRNA
  • ⁇ SEQ_ID_NO 187;RNA;Homo_Sapiens> GII41872532lreflNM_003872.2l Homo sapiens neuropilin 2 (NRP2), transcript variant 2, mRNA
  • PLXNA ⁇ SEQ_ID_NO: 193;RNA;Homo_Sapiens> Gill 13722115lreflNM_025179.31 2 Homo sapiens plexin A2 (PLXNA2), mRNA
  • PLXNB ⁇ SEQ_ID_NO: 194;RNA;Homo_Sapiens> GII194272178lreflNM_002673.4l 1 Homo sapiens plexin Bl (PLXNB 1), transcript variant 1, mRNA
  • GII194272179lreflNM_001130082.1l Homo sapiens plexin B 1 (PLXNB1), transcript variant 2, mRNA
  • GII239582716lreflNM_001031692.2l Homo sapiens leucine rich repeat containing 17 (LRRC17), transcript variant 1, mRNA
  • LIM domain kinase 1 LIMK1
  • LIM domain kinase 2 LIM domain kinase 2
  • transcript variant 1 mRNA
  • KCNC4 Shaw-related subfamily, member 4
  • transcript variant 1 mRNA
  • GII88758575lreflNM_001039574.1l Homo sapiens potassium voltage-gated channel, Shaw-related subfamily, member 4 (KCNC4), transcript variant 3, mRNA
  • KCNE3 Homo sapiens potassium voltage-gated channel, Isk-related family, member 3 (KCNE3), mRNA
  • GCN5 Homo sapiens N-acetyltransferase 8-like (GCN5 -related, putative)
  • FK506 binding protein 1A 12kDa (FKBP1A), transcript variant 12 A, mRNA
  • FK506 binding protein 4 59kDa (FKBP4)
  • LRRK2 leucine-rich repeat kinase 2
  • ubiquitin-conjugating enzyme E2K (UBCl homolog, yeast) (UBE2K), transcript variant 1, mRNA
  • GII163660386lreflNM_001111113.11 Homo sapiens ubiquitin-conjugating enzyme E2K (UBCl homolog, yeast) (UBE2K), transcript variant 3, mRNA
  • WDR33 Homo sapiens WD repeat domain 33 (WDR33), transcript variant 1, mRNA
  • WDR33 Homo sapiens WD repeat domain 33 (WDR33), transcript variant 2, mRNA
  • MYCBP2 Homo sapiens MYC binding protein 2 (MYCBP2), mRNA
  • SEPHS1 selenophosphate synthetase 1
  • HMGB1 Homo sapiens high-mobility group box 1
  • HMGB2 high- mobility group box 2
  • transcript variant 1 mRNA
  • GII194688132lreflNM_001130688.1l Homo sapiens high-mobility group box 2 (HMGB2), transcript variant 2, mRNA
  • GII194688134lreflNM_001130689.1l Homo sapiens high-mobility group box 2 (HMGB2), transcript variant 3, mRNA
  • TRPM7 ⁇ SEQ_ID_NO:235;RNA;Homo_Sapiens> GII148612862lreflNM_017672.3l Homo sapiens transient receptor potential cation channel, subfamily M, member 7 (TRPM7), mRNA
  • THEM4 Homo sapiens thioesterase superfamily member 4 (THEM4), mRNA
  • Homo sapiens solute carrier family 4 sodium bicarbonate cotransporter, member 7 (SLC4A7), mRNA
  • MMP9 matrix metallopeptidase 9 (gelatinase B, 92kDa gelatinase, 92kDa type IV collagenase) (MMP9), mRNA
  • GII295293168lreflNM_001174126.1l Homo sapiens solute carrier family 11 (proton-coupled divalent metal ion transporters), member 2 (SLC11A2), transcript variant 2, mRNA
  • GII295293170lreflNM_001174127.1l Homo sapiens solute carrier family 11 (proton-coupled divalent metal ion transporters), member 2 (SLC11A2), transcript variant 3, mRNA
  • GII295293174lreflNM_001174128.1l Homo sapiens solute carrier family 11 (proton-coupled divalent metal ion transporters), member 2 (SLC11A2), transcript variant 5, mRNA
  • GII295293172lreflNM_001174129.1l Homo sapiens solute carrier family 11 (proton-coupled divalent metal ion transporters), member 2 (SLC11A2), transcript variant 6, mRNA
  • GII295293177lreflNM_001174130.1l Homo sapiens solute carrier family 11 (proton-coupled divalent metal ion transporters), member 2 (SLC11A2), transcript variant 7, mRNA
  • Homo sapiens solute carrier family 11 proton-coupled divalent metal ion transporters, member 2 (SLC11A2), transcript variant 9, non-coding RNA ⁇ SEQ_ID_NO:248;RNA;Homo_Sapiens> GII258613852lreflNR_028457.
  • ATXN3 Homo sapiens ataxin 3
  • transcript variant k non-coding RNA
  • GII258614032lreflNM_001164781.11 Homo sapiens ataxin 3 (ATXN3), transcript variant y, mRNA
  • GII258614028lreflNM_001164779 GII258614028lreflNM_001164779.
  • ATXN3 Homo sapiens ataxin 3
  • transcript variant r transcript variant r
  • GII258614024lreflNM_001164778 GII258614024lreflNM_001164778.
  • ATXN3 Homo sapiens ataxin 3
  • transcript variant o mRNA
  • GII258614018lreflNM_001164774 GII258614018lreflNM_001164774.
  • ATXN3 Homo sapiens ataxin 3
  • transcript variant b transcript variant b
  • ll Homo sapiens ataxin 3 ATXN3
  • transcript variant d non-coding RNA
  • GII258613998lreflNM_001127697.21 Homo sapiens ataxin 3 (ATXN3), transcript variant e, mRNA
  • GII258614022lreflNM_001164776.1l Homo sapiens ataxin 3 (ATXN3), transcript variant g, mRNA
  • GII258614030lreflNM_001164780.1l Homo sapiens ataxin 3 (ATXN3), transcript variant u, mRNA
  • GII189163492lreflNM_001127696.1l Homo sapiens ataxin 3 (ATXN3), transcript variant ad, mRNA
  • GII189491747lreflNM_001128164.1l Homo sapiens ataxin 1 (ATXN1), transcript variant 2, mRNA
  • GII293651612lreflNM_001177387.1l Homo sapiens ataxin 7 (AUXN7), transcript variant SCA7b, mRNA
  • Homo sapiens prion protein PRNP
  • transcript variant 1 mRNA
  • GII122056622lreflNM_001080121.1l Homo sapiens prion protein (PRNP), transcript variant 3, mRNA
  • GII122056624lreflNM_001080122.1l Homo sapiens prion protein (PRNP), transcript variant 4, mRNA
  • GII122056627lreflNM_001080123.1l Homo sapiens prion protein (PRNP), transcript variant 5, mRNA
  • EFNB3 Homo sapiens ephrin-B3 (EFNB3), mRNA
  • EPH receptor A4 EPH receptor A4
  • EFNA5 Homo sapiens ephrin-A5 (EFNA5), mRNA
  • EPH receptor A7 EPH receptor A7
  • EFNB2 Homo sapiens ephrin-B2 (EFNB2), mRNA
  • Table A provides the gi (Genelnfo identifier) and accession numbers for an example of polynucleotide sequences of human mRNA to which the oligonucleotide inhibitors of some of the embodiments of the present invention are directed.
  • inhibition of any one of the genes in Table A is useful in treating and attenuating disease, disorder or injury of the CNS.
  • the 19-mer, 20-mer and 21-mer sense and antisense oligonucleotides useful in the synthesis of double stranded RNA compounds that are utilized in the present invention are selected according to a proprietary algorithm or according to algorithms known in the art.
  • the present invention relates to use of otic pharmaceutical compositions according to the present invention and to methods for the treatment of a subject in need of treatment for a disease, a disorder, or a symptom or a condition associated with the disease or disorder, associated with the expression of a gene selected from the group consisting of SEQ ID NOS: 1-293 comprising administering to the ear of the subject an otic pharmaceutical composition comprising at least one double stranded RNA which reduces or inhibits expression of the gene selected.
  • the subject is a human subject.
  • the double stranded RNA compound is chemically modified according to the embodiments of the present invention.
  • Otic pharmaceutical composition of the invention is useful with any oligonucleotide pair (sense and antisense strands) to a mammalian gene or non-mammalian gene.
  • oligonucleotide compounds administered to the brain, spinal cord and retina according to the non-invasive methods disclosed reach target tissues and cells in the CNS via three different pathways:
  • oligonucleotide compounds will have to pass through the BBB (or, in case of the retina, through the blood-retinal barrier) and only very small amounts of oligonucleotide compounds reaches the CNS through the blood circulation
  • oligonucleotide compound Entering the neurons of the vestibulocochlear nerve. From there the oligonucleotide compound passes through different nerve tracts of the CNS and is delivered to either the retina (via de medial geniculate body (that is common to both the optic nerve tract and the vestibulo cochlear tract and from there to the optic tract) or to different cortical regions (via, for example, the following tract: the cochlear nucleus -> superior olivary nucleus -> inferior colliculus -> medial geniculate nucleus - > auditory cortex). Definitions
  • an “inhibitor” is a compound, which is capable of reducing (partially or fully) the expression of a gene or the activity of the product of such gene to an extent sufficient to achieve a desired biological or physiological effect.
  • the term “inhibitor” as used herein refers to a siRNA inhibitor.
  • a "double stranded RNA inhibitor” is a compound, which is capable of reducing the expression of a gene or the activity of the product of such gene to an extent sufficient to achieve a desired biological or physiological effect.
  • the term as used herein refers to one or more of a siRNA, shRNA, and synthetic shRNA. Inhibition may also be referred to as down- regulation or, for RNAi, silencing.
  • inhibitor refers to reducing the expression of a gene or the activity of the product of such gene to an extent sufficient to achieve a desired biological or physiological effect. Inhibition is either complete or partial.
  • inhibitor of APP gene means inhibition of the gene expression (transcription or translation) or polypeptide activity of one or more of the variants or an SNP (single nucleotide polymorphism) thereof.
  • Central nervous system or “CNS” means the brain, optic nerve, retina and/or spinal cord.
  • central nervous system disorder or "CNS disorder” or “disease of the central nervous system” or “disease of the CNS” means any condition or disease that causes or results in a functional and/or physical deficit in the brain, retina, optic nerve and/or spinal cord or in the cells and tissues which comprise the brain, retina, optic nerve and/or spinal cord.
  • injury of the central nervous system or “injury of the CNS” refers to any and all injury or trauma of the brain, retina, optic nerve and/or spinal cord, including traumatic and non-traumatic injury, that causes or results in an impairment of motor and/or sensory and/or cognitive and/or mental and/or emotional and/or autonomic function.
  • neurodegeneration means the arrest and/or slow down and/or attenuate and/or reverse progression of neurodegeneration.
  • neurodegeneration means the progressive loss of neurons. This includes but is not limited to immediate loss of neurons followed by subsequent loss of connecting or adjacent neurons.
  • Neuron Neuronal cell
  • neural cell including neural progenitor cells and neural stem cells
  • nerve cells i.e., cells that are responsible for conducting nerve impulses from one part of the body to another.
  • Most neurons consist of three distinct portions: a cell body which contains the nucleus, and two different types of cytoplasmic processes: dendrites and axons.
  • Dendrites which are the receiving portion of the neuron, are usually highly branched, thick extensions of the cell body.
  • the axon is typically a single long, thin process that is specialized to conducts nerve impulses away from the cell body to another neuron or muscular or glandular tissue.
  • Axons may have side branches called "axon collaterals.” Axon collaterals and axons may terminate by branching into many fine filaments called telodendria. The distal ends of telodendria are called synaptic end bulbs or axonal terminals, which contain synaptic vesicles that store neurotransmitters. Axons may be surrounded by a multilayered, white, phospholipid, segmented covering called the myelin sheath, which is formed by Schwann cells in the peripheral nervous system and oligodendrocytes in the central nervous system.
  • Axons containing such a covering are "myelinated.”
  • Neurons include sensory (afferent) neurons, which transmit impulses from receptors in the periphery to the brain and spinal cord and from lower to higher centers of the central nervous system.
  • a neuron can also be motor (efferent) neurons which convey impulses from the brain and spinal cord to effectors in the periphery and from higher to lower centers of the central nervous system.
  • Other neurons are association (connecting or interneuron) neurons which carry impulses from sensory neurons to motor neurons and are located within the central nervous system.
  • the processes of afferent and efferent neurons arranged into bundles are called “nerves" when located outside the CNS or fiber tracts if inside the CNS.
  • topical administration or “topical application” is used to mean a local administration of an otic composition to the ear of the subject.
  • otic and “auricular” are used herein interchangeably and generally refer to tissue in and/or around an ear, including the outer ear, the middle ear and the inner ear.
  • ear canal or “external auditory meatus” is used to mean a tube running from the outer ear to the middle ear.
  • tympanic membrane refers to a thin membrane that separates the external ear from the middle ear.
  • phrases such as "pharmaceutical composition” or “otic pharmaceutical composition” or “pharmaceutical formulation” or “otic pharmaceutical formulation” or “pharmaceutical preparation” or “otic pharmaceutical preparation” are used herein interchangebly to generally refer to formulations that are adapted to otic administration and delivery of one or more oligonucleotide active compounds to a CNS, a CNS cell, a group of CNS cells, or a CNS tissue, in an animal or a human.
  • Treatment covers any treatment of a disease or condition of a mammal, particularly a human, and includes: (a) preventing the disease or condition from occurring in a subject which may be predisposed to the disease or condition but has not yet been diagnosed as having it; (b) inhibiting the disease or condition, i.e., arresting or slowing down or postponing its development or progression; (c) relieving and/or ameliorating the disease or condition, i.e., causing regression of the disease or condition and/or the symptoms thereof; or (d) curing the disease or condition, i.e., stopping its development or progression.
  • the population of subjects treated by the methods of the invention includes subjects suffering from the undesirable condition or disease, as well as subjects at risk for development of the condition or disease.
  • the term “pharmaceutically acceptable” means that the components, in addition to the therapeutic agent, comprising the formulation, are suitable for administration to the patient being treated in accordance with the present invention.
  • a “penetration enhancer” or “permeability enhancer” refers to a compound or a combination of compounds that enhance the penetration of a therapeutic oligonucleotide through the skin and/or the tympanic membrane in the ear of an animal or a human.
  • tissue refers to an aggregation of similarly specialized cells united in the performance of a particular function.
  • CNS cells includes one or more of neuronal cells and/or glial cells (e.g. oligodendrocytes, astrocytes, ependymal cells, microglial cells, radial glia cells, or Schwann cells) and include the optic nerve and cells of the retina.
  • glial cells e.g. oligodendrocytes, astrocytes, ependymal cells, microglial cells, radial glia cells, or Schwann cells
  • optic nerve and cells of the retina include the optic nerve and cells of the retina.
  • polynucleotide and “nucleic acid” may be used interchangeably and refer to nucleotide sequences comprising deoxyribonucleic acid (DNA), and ribonucleic acid (RNA).
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • the terms are to be understood to include, as equivalents, analogs of either RNA or DNA made from nucleotide analogs.
  • mRNA sequences are set forth as representing the corresponding genes.
  • Oligonucleotide and “oligomer” are used interchangeably and refer to a deoxyribonucleotide or ribonucleotide sequence from about 2 to about 50 nucleotides. Each DNA or RNA nucleotide may be independently natural or synthetic, and or modified or unmodified. Modifications include changes to the sugar moiety, the base moiety and or the linkages between nucleotides in the oligonucleotide.
  • the compounds of the present invention encompass molecules comprising deoxyribonucleotides, ribonucleotides, modified deoxyribonucleotides, modified ribonucleotides, unconventional moieties and combinations thereof. Oligonucleotide is meant to encompass single stranded molecules including antisense and shRNA, and double stranded molecules including double stranded RNA (dsRNA), siNA, siRNA and miRNA.
  • dsRNA double stranded RNA
  • Substantially complementary refers to complementarity of greater than about 84%, to another sequence.
  • substantially identical refers to identity of greater than about 84%, to another sequence.
  • the present invention provides methods and compositions for inhibiting expression of a target gene in vivo.
  • the method includes topical otic administration of oligoribonucleotides, in particular double stranded RNA compounds (e.g.
  • RNA interference mechanism small interfering RNAs or siRNAs
  • the method can be used to inhibit expression of the gene for treatment of a subject suffering from a disease, disorder or injury related to expression of that gene in CNS tissue or cell.
  • the double stranded RNA molecules or inhibitors of the target gene are used as drugs to treat various CNS pathologies.
  • Nucleotide is meant to encompass deoxyribonucleotides and ribonucleotides, which may be natural or synthetic, and or modified or unmodified. Modifications include changes to the sugar moiety, the base moiety and or the linkages between ribonucleotides in the oligoribonucleotide. As used herein, the term “ribonucleotide” encompasses natural and synthetic, unmodified and modified ribonucleotides. Modifications include changes to the sugar moiety, to the base moiety and/ or to the linkages between ribonucleotides in the oligonucleotide.
  • the nucleotides can be selected from naturally occurring or synthetic modified bases.
  • Naturally occurring bases include adenine, guanine, cytosine, thymine and uracil.
  • Modified bases of nucleotides include inosine, xanthine, hypoxanthine, 2- aminoadenine, 6-methyl, 2- propyl and other alkyl adenines, 5-halo uracil, 5-halo cytosine, 6-aza cytosine and 6-aza thymine, pseudo uracil, 4- thiouracil, 8-halo adenine, 8-aminoadenine, 8-thiol adenine, 8- thiolalkyl adenines, 8-hydroxyl adenine and other 8-substituted adenines, 8-halo guanines, 8- amino guanine, 8-thiol guanine, 8-thioalkyl guanines, 8- hydroxyl guanine
  • the inhibitory oligonucleotide compound comprises unmodified and modified nucleotides and/or unconventional moieties.
  • the compound comprises at least one modified nucleotide selected from the group consisting of a sugar modification, a base modification and an internucleotide linkage modification and may contain DNA, and modified nucleotides such as LNA (locked nucleic acid), ENA (ethylene- bridged nucleic acid), PNA (peptide nucleic acid), arabinoside, phosphonocarboxylate or phosphinocarboxylate nucleotide (PACE nucleotide), mirror nucleotide, or nucleotides with a 6 carbon sugar.
  • LNA locked nucleic acid
  • ENA ethylene- bridged nucleic acid
  • PNA peptide nucleic acid
  • arabinoside phosphonocarboxylate or phosphinocarboxylate nucleotide
  • PACE nucleotide mirror nucleotide
  • nucleotide / oligonucleotide All analogs of, or modifications to, a nucleotide / oligonucleotide are employed with the present invention, provided that said analog or modification does not substantially adversely affect the function of the nucleotide / oligonucleotide.
  • Acceptable modifications include modifications of the sugar moiety, modifications of the base moiety, modifications in the internucleotide linkages and combinations thereof.
  • a sugar modification includes a modification on the 2' moiety of the sugar residue and encompasses amino, fluoro, alkoxy e.g. methoxy, alkyl, amino, fluoro, chloro, bromo, CN, CF, imidazole, carboxylate, thioate, Ci to C 10 lower alkyl, substituted lower alkyl, alkaryl or aralkyl, OCF 3 , OCN; 0-, S-, or N- alkyl; 0-, S, or N-alkenyl; SOCH 3 ; S0 2 CH 3 ; ON0 2 ; N0 2 , N 3 ; heterocycloalkyl; heterocycloalkaryl; amino alky lamino; polyalkylamino or substituted silyl, as, among others, described in European patents EP 0 586 520 Bl or EP 0 618 925 Bl.
  • amino, fluoro, alkoxy e.g. methoxy, alkyl, amino, fluor
  • the double stranded RNA compound comprises at least one ribonucleotide comprising a 2' modification on the sugar moiety ("2' sugar modification").
  • the compound comprises 2'0-alkyl or 2'-fluoro or 2'0-allyl or any other 2' modification, optionally on alternate positions.
  • Other stabilizing modifications are also possible (e.g. terminal modifications).
  • a preferred 2'O-alkyl is 2 ⁇ - methyl (methoxy) sugar modification.
  • the backbone of the oligonucleotides is modified and comprises phosphate-D-ribose entities but may also contain thiophosphate-D-ribose entities, triester, thioate, 2'-5' bridged backbone (also may be referred to as 5'-2'), PACE and the like.
  • non-pairing nucleotide analog means a nucleotide analog which comprises a non-base pairing moiety including but not limited to: 6 des amino adenosine (Nebularine), 4-Me-indole, 3-nitropyrrole, 5-nitroindole, Ds, Pa, N3-Me ribo U, N3-Me riboT, N3-Me dC, N3-Me-dT, Nl-Me-dG, Nl-Me-dA, N3-ethyl-dC, N3-Me dC.
  • the non-base pairing nucleotide analog is a ribonucleotide.
  • analogue of polynucleotides may be prepared wherein the structure of one or more nucleotide is fundamentally altered and better suited as therapeutic or experimental reagents.
  • An example of a nucleotide analogue is a peptide nucleic acid (PNA) wherein the deoxyribose (or ribose) phosphate backbone in DNA (or RNA) is replaced with a polyamide backbone which is similar to that found in peptides.
  • PNA analogues have been shown to be resistant to enzymatic degradation and to have enhanced stability in vivo and in vitro.
  • oligonucleotides include polymer backbones, cyclic backbones, acyclic backbones, thiophosphate-D-ribose backbones, triester backbones, thioate backbones, 2'-5' linked backbone (also lmown as 2'5' nucleotides, or 2'5' ribonucleotides [with 3 ⁇ ]), artificial nucleic acids, morpholino nucleic acids, glycol nucleic acid (GNA), threose nucleic acid (TNA), arabinoside, and mirror nucleoside (for example, beta-L-deoxyribonucleoside instead of beta-D-deoxyribonucleoside).
  • siRNA compounds comprising LNA nucleotides are disclosed in Elmen et al., (NAR 2005, 33(l):439-447).
  • the double stranded RNA compounds are synthesized using one or more inverted nucleotides, for example inverted thymidine or inverted adenine (see, for example, Takei, et al., 2002, JBC 277(26):23800-06).
  • inverted nucleotides for example inverted thymidine or inverted adenine (see, for example, Takei, et al., 2002, JBC 277(26):23800-06).
  • modifications include terminal modifications on the 5' and/or 3' part of the oligonucleotides and are also known as capping moieties.
  • Such terminal modifications are selected from a nucleotide, a modified nucleotide, a lipid, a peptide, a sugar and inverted abasic moiety.
  • a nucleotide is a monomeric unit of nucleic acid, consisting of a ribose or deoxyribose sugar, a phosphate, and a base (adenine, guanine, thymine, or cytosine in DNA; adenine, guanine, uracil, or cytosine in RNA).
  • a modified nucleotide comprises a modification in one or more of the sugar, phosphate and or base.
  • the abasic pseudo- nucleotide lacks a base, and thus is not strictly a nucleotide.
  • the term "capping moiety" as used herein (“ z” ”) includes abasic ribose moiety, abasic deoxyribose moiety, modified abasic ribose and abasic deoxyribose moieties including 2' O alkyl modifications; inverted abasic ribose and abasic deoxyribose moieties and modifications thereof; C6-imino-Pi; a mirror nucleotide including L-DNA and L-RNA; 5'0-Me nucleotide; and nucleotide analogs including 4',5'-methylene nucleotide; 1-( ⁇ - ⁇ - erythrofuranosyl)nucleotide; 4'-thio nucleotide, carbocyclic nucleotide; 5 '-amino -alky
  • capping moieties are abasic ribose or abasic deoxyribose moieties; inverted abasic ribose or abasic deoxyribose moieties; C6-amino-Pi; a mirror nucleotide including L- DNA and L-RNA.
  • Another preferred capping moiety is a C3 non- nucleotide moity derived from propanediol
  • unconventional moiety refers to abasic ribose moiety, an abasic deoxyribose moiety, a deoxyribonucleotide, a modified deoxyribonucleotide, a mirror nucleotide, a non-base pairing nucleotide analog and a nucleotide linked to an adjacent nucleotide by a 2'-5' internucleotide phosphate bond; bridged nucleic acids including LNA and ethylene bridged nucleic acids.
  • a preferred unconventional moiety is an abasic ribose moiety, an abasic deoxyribose moiety, a deoxyribonucleotide, a mirror nucleotide, and a nucleotide linked to an adjacent nucleotide by a 2'-5' internucleotide phosphate bond.
  • Abasic deoxyribose moiety includes for example abasic deoxyribose-3'-phosphate; 1,2- dideoxy-D-ribofuranose-3-phosphate; 1 ,4-anhydro-2-deoxy-D-ribitol-3-phosphate.
  • Inverted abasic deoxyribose moiety includes inverted deoxyriboabasic; 3', 5' inverted deoxyabasic 5'- phosphate.
  • a "mirror nucleotide” is a nucleotide with reversed chirality to the naturally occurring or commonly employed nucleotide, i.e., a mirror image (L- nucleotide) of the naturally occurring (D-nucleotide), also referred to as L-RNA in the case of a mirror ribonucleotide, and "aptmer".
  • the nucleotide can be a ribonucleotide or a deoxyribonucleotide and my further comprise at least one sugar, base and or backbone modification. See US Patent No. 6,586,238. Also, US Patent No. 6,602,858 discloses nucleic acid catalysts comprising at least one L- nucleotide substitution.
  • Mirror nucleotide includes for example L-DNA (L- deoxyriboadenosine-3'-phosphate (mirror dA); L-deoxyribocytidine-3'-phosphate (mirror dC); L-deoxyriboguanosine-3'-phosphate (mirror dG); L-deoxyribothymidine-3'-phosphate (mirror image dT)) and L-RNA (L-riboadenosine-3'-phosphate (mirror rA); L-ribocytidine- 3'-phosphate (mirror rC); L-riboguanosine-3'-phosphate (mirror rG); L-ribouracil-3'- phosphate (mirror rU).
  • L-DNA L- deoxyriboadenosine-3'-phosphate
  • mirror dC L-deoxyribocytidine-3'-phosphate
  • mirror dG L-deoxy
  • Modified deoxyribonucleotide includes, for example 5'OMe DNA (5-methyl- deoxyriboguanosine-3'-phosphate) which may be useful as a nucleotide in the 5' terminal position (position number 1); PACE (deoxyriboadenine 3' phosphonoacetate, deoxyribocytidine 3' phosphonoacetate, deoxyriboguanosine 3' phosphonoacetate, deoxyribothymidine 3' phosphonoacetate).
  • 5'OMe DNA 5-methyl- deoxyriboguanosine-3'-phosphate
  • PACE deoxyriboadenine 3' phosphonoacetate
  • deoxyribocytidine 3' phosphonoacetate deoxyriboguanosine 3' phosphonoacetate
  • deoxyribothymidine 3' phosphonoacetate deoxyribothymidine 3' phosphonoacetate
  • Bridged nucleic acids include LNA (2'-0, 4'-C-methylene bridged Nucleic Acid adenosine 3' monophosphate, 2'-0,4'-C-methylene bridged Nucleic Acid 5-methyl-cytidine 3' monophosphate, 2'-0,4'-C-methylene bridged Nucleic Acid guanosine 3' monophosphate, 5- methyl-uridine (or thymidine) 3' monophosphate); and ENA (2'-0,4'-C-ethylene bridged Nucleic Acid adenosine 3' monophosphate, 2'-0,4'-C-ethylene bridged Nucleic Acid 5- methyl-cytidine 3' monophosphate, 2'-0,4'-C-ethylene bridged Nucleic Acid guanosine 3' monophosphate, 5-methyl-uridine (or thymidine) 3' monophosphate).
  • LNA 2'-0, 4'-C-methylene bridged Nucleic Acid aden
  • the present invention provides inhibitory oligonucleotide compounds comprising unmodified and modified nucleotides.
  • the compound comprises at least one modified nucleotide selected from the group consisting of a sugar modification, a base modification and an internucleotide linkage modification and may contain DNA, and modified nucleotides such as LNA (locked nucleic acid) including ENA (ethylene-bridged nucleic acid; PNA (peptide nucleic acid); arabinoside; PACE (phosphonoacetate and derivatives thereof), mirror nucleotide, or nucleotides with a six-carbon sugar.
  • LNA locked nucleic acid
  • ENA ethylene-bridged nucleic acid
  • PNA peptide nucleic acid
  • arabinoside PACE (phosphonoacetate and derivatives thereof), mirror nucleotide, or nucleotides with a six-carbon sugar.
  • PCT applications have recently been published that relate to the RNAi phenomenon. These include: PCT publication WO 00/44895; PCT publication WO 00/49035; PCT publication WO 00/63364; PCT publication WO 01/36641 ; PCT publication WO 01/36646; PCT publication WO 99/32619; PCT publication WO 00/44914; PCT publication WO 01/29058; and PCT publication WO 01/75164.
  • RNA interference is based on the ability of dsRNA species to enter a cytoplasmic protein complex, where it is then targeted to the complementary cellular RNA and specifically degrade it.
  • the RNA interference response features an endonuclease complex containing a siRNA, commonly referred to as an RNA-induced silencing complex (RISC), which mediates cleavage of single- stranded RNA having a sequence complementary to the antisense strand of the siRNA duplex. Cleavage of the target RNA may take place in the middle of the region complementary to the antisense strand of the siRNA duplex (Elbashir et al., Genes Dev., 2001, 15(2): 188-200).
  • RISC RNA-induced silencing complex
  • dsRNAs are digested into short (17-29 bp) dsRNA fragments (also referred to as short inhibitory RNAs, "siRNAs") by type III RNAses (DICER, DROSHA, etc.; Bernstein et al., Nature, 2001, 409(6818):363-6; Lee et al., Nature, 2003, 425(6956):415-9).
  • the RISC protein complex recognizes these fragments and complementary mRNA. The whole process is culminated by endonuclease cleavage of target mRNA (McManus & Sharp, Nature Rev Genet, 2002, 3(10):737-47; Paddison & Hannon, Curr Opin Mol Ther.
  • siRNA corresponding to known genes has been widely reported; see for example Ui-Tei et al., J Biomed Biotechnol. 2006; 2006: 65052; Chalk et al., BBRC. 2004, 319(1): 264-74; Sioud & Leirdal, Met. Mol Biol.; 2004, 252:457-69; Levenkova et al., Bio inform. 2004, 20(3):430-2; Ui-Tei et al., Nuc. Acid Res. 2004, 32(3):936-48.
  • a siRNA is a double- stranded RNA (dsRNA) which down-regulates or silences (i.e. fully or partially inhibits) the expression of an endogenous or exogenous gene/mRNA.
  • RNA interference is based on the ability of certain dsRNA species to enter a specific protein complex, where they are then targeted to complementary cellular RNA (i.e. mRNA), which they specifically degrade or cleave.
  • mRNA complementary cellular RNA
  • the RNA interference response features an endonuclease complex containing siRNA, commonly referred to as an RNA-induced silencing complex (RISC), which mediates cleavage of single- stranded RNA having a sequence complementary to the antisense strand of the siRNA duplex.
  • RISC RNA-induced silencing complex
  • Cleavage of the target RNA may take place in the middle of the region complementary to the antisense strand of the siRNA duplex (Elbashir, et al., Genes Dev., 2001, 15: 188).
  • longer dsRNAs are digested into short (17-29 bp) dsRNA fragments (also referred to as short inhibitory RNAs or "siRNAs") by type III RNAses (DICER, DROSHA, etc., see Bernstein et al., Nature, 2001, 409:363-6 and Lee et al., Nature, 2003, 425:415-9).
  • DIER type III RNAses
  • siRNA can be effective in vivo in mammals including humans. Specifically, Bitko et al., showed that specific siRNAs directed against the respiratory syncytial virus (RSV) nucleocapsid N gene are effective in treating mice when administered intranasally (Nat. Med. 2005, l l(l):50-55). For reviews of therapeutic applications of siRNAs see for example Barik (Mol. Med 2005, 83: 764-773) and Chakraborty (Current Drug Targets 2007 8(3):469-82).
  • RSV respiratory syncytial virus
  • siRNA corresponding to known genes has been widely reported; (see for example Ui-Tei et al., 2006. J Biomed Biotechnol. 2006:65052; Chalk et al., 2004. BBRC. 319(1): 264-74; Sioud & Leirdal, 2004. Met. Mol Biol. 252:457-69; Levenkova et al., 2004, Bioinform. 20(3):430-2; Ui-Tei et al., 2004. NAR 32(3):936-48).
  • Holen et al (2003, NAR, 31(9):2401-2407) report that an siRNA having small numbers of 2'-0-methyl modified nucleosides showed good activity compared to wild type but that the activity decreased as the numbers of 2'-0- methyl modified nucleosides was increased.
  • Chiu and Rana (2003, RNA, 9: 1034-1048) teach that incorporation of 2'-0-methyl modified nucleosides in the sense or antisense strand (fully modified strands) severely reduced siRNA activity relative to unmodified siRNA.
  • Otic pharmaceutical compositions of the invention are prepared using any chemically modified or non-modified double stranded RNA oligonucleotide compound.
  • siRNA which are Dicer substrates or asymmetric siRNA may be used with the invention.
  • Double stranded RNA oligonucleotide compounds used in the present invention encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an mammal, including a human, is capable of treating diseases, disorders and injury of the CNS.
  • pharmaceutically acceptable salts refers to physiologically and pharmaceutically acceptable salts, i. e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto.
  • otic pharmaceutical compositions of the invention are prepared using double stranded RNA compounds that are chemically and or structurally modified according to one of the following modifications set forth in Structures disclsoed herein or as tandem siRNA or RNAstar (see below).
  • nucleotide positions are numbered from 1 to 19 or 1 to 21 or 1 to 23 and are counted from the 5' end of the antisense or sense oligonucleotides.
  • position 1 on (N)x refers to the 5' terminal nucleotide on the antisense oligonucleotide strand
  • position 1 on (N')y refers to the 5' terminal nucleotide on the sense oligonucleotide strand.
  • the present invention provides an otic pharmaceutical composition comprising a therapeutically effective amount of a double stranded RNA molecule set forth as Structure (A):
  • N and N' is a nucleotide which may be unmodified or modified, or an unconventional moiety
  • each of (N) x and (N') y is an oligonucleotide in which each consecutive N or N' is joined to the next N or N' by a covalent bond; wherein each of x and y is an integer between 18 and 40;
  • each of Z and Z' may be present or absent, but if present is 1-5 consecutive nucleotides or no n- nucleotide moieties covalently attached at the 3' terminus of the strand in which it is present;
  • z may be present or absent, but if present is a capping moiety covalently attached at the 5' terminus of (N')y;
  • each of x and y is independently an integer from 18 to 40;
  • sequence of (N')y has complementary to the sequence of (N)x; and wherein (N)x includes an antisense sequence to mRNA of a gene upregulated in a disease, disorder or injury of the CNS.
  • (N)x comprises an antisense that is complementary to about 18 to about 40 consecutive ribonucleotides in a target mRNA set forth in any one of SEQ ID NOS: 1-293.
  • (N)x and (N')y are oligonucleotide pairs provided in PCT Patent Publication Nos. WO 2006/023544, WO 2007/084684, WO 2008/050329, WO 2007/141796, WO 2009/044392, WO 2008/106102, WO 2008/152636, WO 2009/001359, WO/2009/090639 assigned to the assignee of the present invention and incorporated herein by reference in their entirety.
  • the covalent bond joining each consecutive N or N' is a phosphodiester bond.
  • Z and Z' are absent. In other embodiments one of Z or Z' is present.
  • the 5' terminal nucleotide of the antisense strand (position 1 of the antisense strand) is mismatched to the target mRNA. In some embodiments the 5' terminal nucleotide of the antisense strand is a modified riboadenosine or a modified ribouridine. In some embodiments each of (N) x and (N') y is independently phosphorylated or non- phosphorylated at the 3' and 5' termini.
  • each N at the 5' and 3' termini of (N) x is modified; and each N' at the 5' and 3' termini of (N') y is unmodified.
  • each N at the 5' and 3' termini of (N) x is unmodified; and each N' at the 5' and 3' termini of (N') y is modified.
  • x and y 19, and the double stranded RNA compound is modified such that a 2'OMe sugar modified ribonucleotide (2'OMe) is present in nuclease snestive positions.
  • at least one pyrimidine comprises a 2'OMe sugar modification.
  • all pyrimidines comprise a 2'OMe sugar modification.
  • 2'OMe sugar modified ribonucleotides are present in the first, third, fifth, seventh, ninth, eleventh, thirteenth, fifteenth, seventeenth and nineteenth positions of the antisense strand (N) x
  • a 2'-OMe sugar modified ribonucleotide is present in the second, fourth, sixth, eighth, tenth, twelfth, fourteenth, sixteenth and eighteenth positions of the sense strand (N') y .
  • nucleotides are unmodified or (N)x comprises alternating 2'OMe sugar modified ribonucleotides and unmodified ribonucleotides; and the ribonucleotide located at the middle position of (N)x being modified or unmodified preferably unmodified.
  • (N')y comprises unmodified ribonucleotides further comprising one modified nucleotide at a terminal or penultimate position, wherein the modified nucleotide is selected from the group consisting of a mirror nucleotide, a bicyclic nucleotide, a 2'-sugar modified nucleotide, an altritol nucleotide, or a nucleotide joined to an adjacent nucleotide by an internucleotide linkage selected from a 2'-5' phosphodiester bond, a P-alkoxy linkage or a PACE linkage;
  • each modified ribonucleotide is a 2'OMe sugar modified ribonucleotide and the ribonucleotide located at the middle of (N)x is unmodified.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 1, 3, 5, 7, 9, 11, 13, 15, 17 and 19.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 6, 8, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 5.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 8, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 6.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 6, 8, 11, 13, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 15.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 6, 8, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 14.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 1, 2, 3, 7, 9, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 5.
  • (N)x comprises 2'0 Memodified ribonucleotides at positions 1, 2, 3, 5, 7, 9, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 6.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 1, 2, 3, 5, 7, 9, 11, 13, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 15.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 1, 2, 3, 5, 7, 9, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 14.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 6, 7, 9, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 5.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 1, 2, 4, 6, 7, 9, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 5 .
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 6, 8, 11, 13, 14, 16, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 15.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 1, 2, 3, 5, 7, 9, 11, 13, 14, 16, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 15.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 6, 8, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 7.
  • (N)x comprises 2'0-Me modified ribonucleotides at positions 2, 4, 6, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 8.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 6, 8, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 9.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 6, 8, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 10.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 6, 8, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 11.
  • (N)x comprises 2'OMe sugar modified ribonucleotides at positions 2, 4, 6, 8, 11, 13, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 12.
  • (N)x comprises 2'O-Me modified ribonucleotides at positions 2, 4, 6, 8, 11, 15, 17 and 19 and may further comprise at least one abasic or inverted abasic unconventional moiety for example in position 13.
  • (N)x comprises at least one nucleotide mismatch relative to the target mRNA. In certain preferred embodiments, (N)x comprises a single nucleotide mismatch in position 1, relative to the target RNA. In one embodiment at least two nucleotides at either or both the 5' and 3' termini of (N')y are joined by a 2' -5' phosphodiester bond.
  • an additional nucleotide located in the middle position of (N)y is a 2'OMe sugar modified ribonucleotide.
  • nucleotides alternate between 2'OMe sugar modified ribonucleotides and unmodified ribonucleotides
  • nucleotides in (N)x are joined by three 2'-5' phosphodiester bonds and the 5' terminal nucleotide or two or three consecutive nucleotides at the 5' terminus comprise 3 '-0- methyl (3 OMe) modifications.
  • the following positions comprise an abasic or inverted abasic: positions 1 and 16-19, positions 15-19, positions 1-2 and 17-19, positions 1-3 and 18-19, positions 1-4 and 19 and positions 1-5.
  • (N')y may further comprise at least one LNA nucleotide.
  • the mirror nucleotide is an L-DNA nucleotide.
  • the L-DNA is L-deoxyribocytidine.
  • (N')y comprises L-DNA at position 18.
  • (N')y comprises L-DNA at positions 17 and 18.
  • (N')y comprises L-DNA substitutions at positions 2 and at one or both of positions 17 and 18.
  • (N')y further comprises a 5' terminal cap nucleotide such as 5 '-0- methyl DNA or an abasic or inverted abasic moiety as an overhang.
  • N')y comprises a DNA at position 15 and L-DNA at one or both of positions 17 and 18.
  • position 2 may further comprise an L-DNA or an abasic unconventional moiety.
  • consecutive ribonucleotides at the 3' terminus are linked by 2' -5' internucleotide linkages.
  • four consecutive nucleotides at the 3' terminus of (N')y are joined by three 2' -5' phosphodiester bonds, wherein one or more of the 2'-5' nucleotides which form the 2'-5' phosphodiester bonds further comprises a 3 '-0- methyl sugar modification.
  • the 3' terminal nucleotide of (N')y is a 2'OMe sugar modified ribonucleotide.
  • nucleotide forming the 2'-5' internucleotide bond comprises a 3' deoxyribose nucleotide or a 3' methoxy nucleotide.
  • nucleotides at positions 17 and 18 in (N')y are joined by a 2'-5' internucleotide bond.
  • nucleotides at positions 16-17, 17-18, or 16- 18 in (N')y are joined by a 2' -5' internucleotide bond.
  • (N')y comprises an L-DNA at position 2 and 2'-5' internucleotide bonds at positions 16-17, 17-18, or 16-18. In certain embodiments (N')y comprises 2'-5' internucleotide bonds at positions 16-17, 17-18, or 16-18 and a 5' terminal cap nucleotide.
  • nucleotide in (N')y 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive nucleotides at either terminus or 2-8 modified nucleotides at each of the 5' and 3' termini are independently mirror nucleotides.
  • the mirror nucleotide is an L-ribonucleotide. In other embodiments the mirror nucleotide is an L-deoxyribonucleotide.
  • the mirror nucleotide may further be modified at the sugar or base moiety or in an internucleotide linkage.
  • the 3' terminal nucleotide or two or three consecutive nucleotides at the 3' terminus of (N')y are L-deoxyribonucleotides.
  • consecutive ribonucleotides at either terminus or 2-8 modified nucleotides at each of the 5' and 3' termini are independently 2' sugar modified nucleotides.
  • the 2' sugar modification comprises the presence of an amino, a fhioro, an alkoxy or an alkyl moiety.
  • the 2' sugar modification comprises a methoxy moiety (2'-OMe).
  • three, four or five consecutive nucleotides at the 5' terminus of (N')y comprise the 2'-OMe modification.
  • three consecutive nucleotides at the 3' terminus of (N')y are 2'OMe sugar modified ribonucleotides.
  • ribonucleotides at either or 2-8 modified nucleotides at each of the 5' and 3' termini are independently bicyclic nucleotides.
  • the bicyclic nucleotide is a locked nucleic acid (LNA) or a species of LNA, e.g. 2'-0, 4'-C-ethylene-bridged nucleic acid (ENA) is a species of LNA.
  • (N')y comprises modified nucleotides at the 5' terminus or at both the 3' and 5' termini.
  • At least two nucleotides at either or both the 5' and 3' termini of (N')y are joined by P-ethoxy backbone modifications.
  • three consecutive nucleotides at the 3' terminus or at the 5' terminus of (N')y are joined by two P-ethoxy backbone modifications.
  • consecutive ribonucleotides at each of the 5' and 3' termini are independently mirror nucleotides, nucleotides joined by 2'-5' phosphodiester bond, 2' sugar modified nucleotides or bicyclic nucleotide.
  • the modification at the 5' and 3' termini of (N')y is identical.
  • four consecutive nucleotides at the 5' terminus of (N')y are joined by three 2' -5' phosphodiester bonds and three consecutive nucleotides at the 3' terminus of (N')y are joined by two 2'-5' phosphodiester bonds.
  • the modification at the 5' terminus of (N')y is different from the modification at the 3' terminus of (N')y.
  • the modified nucleotides at the 5' terminus of (N')y are mirror nucleotides and the modified nucleotides at the 3' terminus of (N')y are joined by 2'-5' phosphodiester bond.
  • three consecutive nucleotides at the 5' terminus of (N')y are LNA nucleotides and three consecutive nucleotides at the 3' terminus of (N')y are joined by two 2'-5' phosphodiester bonds.
  • (N)x the nucleotides alternate between 2'OMe sugar modified ribonucleotides and unmodified ribonucleotides, and the ribonucleotide located at the middle of (N)x being unmodified, or the ribonucleotides in (N)x being unmodified.
  • five consecutive nucleotides at the 5' terminus of (N')y comprise the 2'OMe sugar modification and two consecutive nucleotides at the 3' terminus of (N')y are L- DNA.
  • the 5' or 3' terminal nucleotide or 2, 3, 4, 5 or 6 consecutive nucleotides at either termini or 1-4 modified nucleotides at each of the 5' and 3' termini are independently phosphonocarboxylate or phosphinocarboxylate nucleotides (PACE nucleotides).
  • PACE nucleotides are deoxyribonucleotides.
  • 1 or 2 consecutive nucleotides at each of the 5' and 3' termini are PACE nucleotides.
  • (N)x comprises unmodified ribonucleotides in which two consecutive nucleotides linked by one 2'-5' internucleotide linkage at the 3' terminus; and
  • (N')y comprises unmodified ribonucleotides in which two consecutive nucleotides are linked by one 2'-5' internucleotide linkage at the 5' terminus.
  • (N)x comprises unmodified ribonucleotides in which three consecutive nucleotides at the 3' terminus are joined together by two 2'-5' phosphodiester bonds; and
  • (N')y comprises unmodified ribonucleotides in which four consecutive nucleotides at the 5' terminus are joined together by three 2'-5' phosphodiester bonds.
  • 2, 3, 4, 5, 6, 7, consecutive ribonucleotides starting at the ultimate or penultimate position of the 3' terminus of (N)x and 2, 3, 4, 5, 6, 7, consecutive ribonucleotides starting at the ultimate or penultimate position of the 5' terminus of (N')y are linked by 2'-5' internucleotide linkages.
  • nucleotides at the 5' terminus of (N')y are joined by three 2'-5' phosphodiester bonds and three consecutive nucleotides at the 3' terminus of (N')x are joined by two 2'-5' phosphodiester bonds.
  • Three nucleotides at the 5' terminus of (N')y and two nucleotides at the 3' terminus of (N')x may also comprise 3'-0- methyl modifications.
  • 2, 3, 4, consecutive nucleotides starting at the ultimate or penultimate position of the 3' terminus of (N)x and 2, 3, 4 consecutive ribonucleotides starting at the ultimate or penultimate position of the 5' terminus of (N')y are independently mirror nucleotides.
  • the mirror is an L-ribonucleotide.
  • the mirror nucleotide is L-deoxyribonucleotide.
  • 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive ribonucleotides starting at the ultimate or penultimate position of the 3' terminus of (N)x and 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive ribonucleotides starting at the ultimate or penultimate position of the 5' terminus of (N')y are independently 2' sugar modified nucleotides.
  • the 2' sugar modification comprises an amino, a fluoro, an alkoxy or an alkyl moiety.
  • the 2' sugar modification comprises a methoxy moiety (2'- OMe).
  • five consecutive nucleotides at the 5' terminus of (N')y comprise the 2'OMe modification and five consecutive nucleotides at the 3' terminus of (N')x comprise the 2'OMe modification.
  • five to ten consecutive nucleotides at the 5' terminus of (N')y comprise the 2'OMe modification and five consecutive nucleotides at the 3' terminus of (N')x comprise the 2'OMe modification.
  • thirteen consecutive nucleotides at the 5' terminus of (N')y comprise the 2'OMe modification and five consecutive nucleotides at the 3' terminus of (N')x comprise the 2'OMe modification.
  • 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive ribonucleotides starting at the ultimate or penultimate position of the 3' terminus of (N)x and 2, 3, 4, 5, 6, 7, 8 , 9, 10, 11, 12, 13 or 14 consecutive ribonucleotides starting at the ultimate or penultimate position of the 5' terminus of (N')y are independently a bicyclic nucleotide.
  • the bicyclic nucleotide is a locked nucleic acid (LNA) such as a 2'-0, 4'-C- ethylene-bridged nucleic acid (EN A).
  • (N')y comprises a modified nucleotide selected from a bicyclic nucleotide, a 2' sugar modified nucleotide, a mirror nucleotide, an altritol nucleotide or a nucleotide joined to an adjacent nucleotide by an internucleotide linkage selected from a 2'- 5' phosphodiester bond, a P-alkoxy linkage or a PACE linkage;
  • (N)x comprises a modified nucleotide selected from a bicyclic nucleotide, a 2' sugar modified nucleotide, a mirror nucleotide, an altritol nucleotide or a nucleotide joined to an adjacent nucleotide by an internucleotide linkage selected from a 2'- 5' phosphodiester bond, a P-alkoxy linkage or a PACE linkage;
  • each of the 3' and 5' termini of the same strand comprises a modified nucleotide
  • the modification at the 5' and 3' termini is identical.
  • the modification at the 5' terminus is different from the modification at the 3' terminus of the same strand.
  • the modified nucleotides at the 5' terminus are mirror nucleotides and the modified nucleotides at the 3' terminus of the same strand are joined by 2'-5' phosphodiester bond.
  • five consecutive nucleotides at the 5' terminus of (N')y comprise the 2'OMe modification and two consecutive nucleotides at the 3' terminus of (N')y are L- DNA.
  • the compound may further comprise five consecutive 2'OMe sugar modified nucleotides at the 3' terminus of (N')x.
  • the modified nucleotides in (N)x are different from the modified nucleotides in (N')y.
  • the modified nucleotides in (N)x are 2' sugar modified nucleotides and the modified nucleotides in (N')y are nucleotides linked by 2'-5' internucleotide linkages.
  • the modified nucleotides in (N)x are mirror nucleotides and the modified nucleotides in (N')y are nucleotides linked by 2'-5' internucleotide linkages.
  • the modified nucleotides in (N)x are nucleotides linked by 2'-5' internucleotide linkages and the modified nucleotides in (N')y are mirror nucleotides.
  • 2, 3, or 4 consecutive ribonucleotides starting at the ultimate or penultimate position of the 5' terminus of (N)x, preferably starting at the 5' penultimate position, and 2, 3, 4, 5, or 6, consecutive ribonucleotides starting at the ultimate or penultimate position of the 3' terminus of (N')y are linked by 2'-5' internucleotide linkages.
  • 2, 3, or 4 consecutive nucleotides starting at the ultimate or penultimate position of the 5' terminus of (N)x, preferably starting at the 5' penultimate position, and 2, 3, 4, 5, or 6 consecutive nucleotides starting at the ultimate or penultimate position of the 3' terminus of (N')y are independently mirror nucleotides.
  • the mirror is an L-ribonucleotide.
  • the mirror nucleotide is L- deoxyribo nucleotide .
  • 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive ribonucleotides starting at the ultimate or penultimate position of the 5' terminus of (N)x, preferably starting at the 5' penultimate position, and 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive ribonucleotides starting at the ultimate or penultimate position of the 3' terminus of (N')y are independently 2' sugar modified nucleotides.
  • the 2' sugar modification comprises an amino, a fluoro, an alkoxy or an alkyl moiety.
  • the 2' sugar modification comprises a methoxy moiety (2'-OMe).
  • 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive ribonucleotides starting at the ultimate or penultimate position of the 5' terminus of (N)x, preferably starting at the 5' penultimate position, and 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive ribonucleotides starting at the ultimate or penultimate position of the 3' terminus of (N')y are independently a bicyclic nucleotide.
  • the bicyclic nucleotide is a locked nucleic acid (LNA) such as a 2'-0, 4'-C-ethylene-bridged nucleic acid (ENA).
  • LNA locked nucleic acid
  • ENA 2'-0, 4'-C-ethylene-bridged nucleic acid
  • N' comprises modified nucleotides selected from a bicyclic nucleotide, a 2' sugar modified nucleotide, a mirror nucleotide, an altritol nucleotide, a nucleotide joined to an adjacent nucleotide by an internucleotide linkage selected from a 2'- 5' phosphodiester bond, a P-alkoxy linkage or a PACE linkage at the 3' terminus or at each of the 3' and 5' termini.
  • (N)x comprises a modified nucleotide selected from a bicyclic nucleotide, a 2' sugar modified nucleotide, a mirror nucleotide, an altritol nucleotide, or a nucleotide joined to an adjacent nucleotide by an internucleotide linkage selected from a 2'- 5' phosphodiester bond, a P-alkoxy linkage or a PACE linkage at the 5' terminus or at each of the 3' and 5' termini.
  • both 3' and 5' termini of the same strand comprise a modified nucleotide
  • the modification at the 5' and 3' termini is identical.
  • the modification at the 5' terminus is different from the modification at the 3' terminus of the same strand.
  • the modified nucleotides at the 5' terminus are mirror nucleotides and the modified nucleotides at the 3' terminus of the same strand are joined by 2'-5' phosphodiester bond.
  • the modified nucleotides in (N)x are different from the modified nucleotides in (N')y.
  • the modified nucleotides in (N)x are 2' sugar modified nucleotides and the modified nucleotides in (N')y are nucleotides linked by 2'-5' internucleotide linkages.
  • the modified nucleotides in (N)x are mirror nucleotides and the modified nucleotides in (N')y are nucleotides linked by 2'-5' internucleotide linkages.
  • the modified nucleotides in (N)x are nucleotides linked by 2'-5' internucleotide linkages and the modified nucleotides in (N')y are mirror nucleotides.
  • the modification at the 5' and 3' termini is identical.
  • the modification at the 5' terminus is different from the modification at the 3' terminus of the same strand.
  • the modified nucleotides at the 5' terminus are mirror nucleotides and the modified nucleotides at the 3' terminus of the same strand are joined by 2'-5' phosphodiester bond.
  • the modified nucleotides in (N)x are different from the modified nucleotides in (N')y.
  • the modified nucleotides in (N)x are 2' sugar modified nucleotides and the modified nucleotides in (N')y are nucleotides linked by 2'-5' internucleotide linkages.
  • the modified nucleotides in (N)x are mirror nucleotides and the modified nucleotides in (N')y are nucleotides linked by 2'-5' internucleotide linkages.
  • the modified nucleotides in (N)x are nucleotides linked by 2'-5' internucleotide linkages and the modified nucleotides in (N')y are mirror nucleotides.
  • 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive ribonucleotides independently beginning at the ultimate or penultimate position of the 5' termini of (N)x and (N')y are independently 2' sugar modified nucleotides.
  • the 2' sugar modification comprises an amino, a fluoro, an alkoxy or an alkyl moiety.
  • the 2' sugar modification comprises a methoxy moiety (2'-OMe).
  • the consecutive modified nucleotides preferably begin at the penultimate position of the 5' terminus of (N)x.
  • five consecutive ribonucleotides at the 5' terminus of (N')y comprise a 2'OMe modification and one ribonucleotide at the 5' penultimate position of (N')x comprises a 2'OMe modification.
  • five consecutive ribonucleotides at the 5' terminus of (N')y comprise a 2'OMe modification and two consecutive ribonucleotides at the 5' terminal position of (N')x comprise a 2'OMe modification.
  • (N')y comprises a modified nucleotide selected from a bicyclic nucleotide, a 2' sugar modified nucleotide, a mirror nucleotide, an altritol nucleotide, or a nucleotide joined to an adjacent nucleotide by an internucleotide linkage selected from a 2'- 5' phosphodiester bond, a P-alkoxy linkage or a PACE linkage at the 5' terminus or at each of the 3' and 5' termini.
  • (N)x comprises a modified nucleotide selected from a bicyclic nucleotide, a 2' sugar modified nucleotide, a mirror nucleotide, an altritol nucleotide, or a nucleotide joined to an adjacent nucleotide by an internucleotide linkage selected from a 2'- 5' phosphodiester bond, a P-alkoxy linkage or a PACE linkage at the 5' terminus or at each of the 3' and 5' termini.
  • each of 3' and 5' termini of the same strand comprise a modified nucleotide
  • the modification at the 5' and 3' termini is identical.
  • the modification at the 5' terminus is different from the modification at the 3' terminus of the same strand.
  • the modified nucleotides at the 5' terminus are mirror nucleotides and the modified nucleotides at the 3' terminus of the same strand are joined by 2'-5' phosphodiester bond.
  • the modified nucleotides in (N)x are different from the modified nucleotides in (N')y.
  • the modified nucleotides in (N)x are 2' sugar modified nucleotides and the modified nucleotides in (N')y are nucleotides linked by 2' -5' internucleotide linkages.
  • the modified nucleotides in (N)x are mirror nucleotides and the modified nucleotides in (N')y are nucleotides linked by 2'-5' internucleotide linkages.
  • the modified nucleotides in (N)x are nucleotides linked by 2' -5' internucleotide linkages and the modified nucleotides in (N')y are mirror nucleotides.
  • 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive ribonucleotides independently beginning at the ultimate or penultimate position of the 3' terminus or the 5' terminus or 2-8 consecutive nucleotides at each of 5' and 3' termini of (N')y are independently 2' sugar modified nucleotides, bicyclic nucleotides, mirror nucleotides, altritol nucleotides or nucleotides joined to an adjacent nucleotide by a 2'-5' phosphodiester bond
  • 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive internal ribonucleotides in (N)x are independently 2' sugar modified nucleotides, bicyclic nucleotides, mirror nucleotides, altritol nucleotides or nucleotides joined to an adjacent nucleotide by a 2'-5' phosphodiester bond.
  • each of 3' and 5' termini of the same strand comprises a modified nucleotide
  • the modification at the 5' and 3' termini is identical.
  • the modification at the 5' terminus is different from the modification at the 3' terminus of the same strand.
  • the modified nucleotides at the 5' terminus are mirror nucleotides and the modified nucleotides at the 3' terminus of the same strand are joined by 2'-5' phosphodiester bond.
  • the modified nucleotides in (N)x are different from the modified nucleotides in (N')y.
  • the modified nucleotides in (N)x are 2' sugar modified nucleotides and the modified nucleotides in (N')y are nucleotides linked by 2'-5' internucleotide linkages.
  • the modified nucleotides in (N)x are mirror nucleotides and the modified nucleotides in (N')y are nucleotides linked by 2'-5' internucleotide linkages.
  • the compounds of the invention comprise modified ribonucleotides in alternating positions wherein each N at the 5' and 3' termini of (N)x is modified in its sugar residue and the middle ribonucleotide is not modified, e.g. ribonucleotide in position 10 in a 19-mer strand, position 11 in a 21- mer and position 12 in a 23-mer strand.
  • neither (N)x nor (N')y are phosphorylated at the 3' and 5' termini. In other embodiments either or both (N)x and (N')y are phosphorylated at the 3' termini. In yet another embodiment, either or both (N)x and (N')y are phosphorylated at the 3' termini using non-cleavable phosphate groups. In yet another embodiment, either or both (N)x and (N')y are phosphorylated at the terminal 5' termini position using cleavable or non-cleavable phosphate groups.
  • the double stranded RNA compounds are blunt ended and are non-phosphorylated at the termini; however, comparative experiments have shown that double stranded RNA compounds phosphorylated at one or both of the 3 '-termini have similar activity in vivo compared to the non-phosphorylated compounds.
  • the double stranded RNA compound is blunt ended, for example wherein both Z and Z' are absent.
  • the compound comprises at least one 3' overhang, wherein at least one of Z or Z' is present.
  • Z and Z' independently comprises one or more covalently linked modified or non-modified nucleotides, for example inverted dT or dA; dT, LNA, mirror nucleotide and the like.
  • each of Z and Z' are independently selected from dT and dTdT.
  • Double stranded RNA in which Z and/or Z' is present have similar activity and stability as double stranded RNA in which Z and Z' are absent.
  • the double stranded RNA compound comprises one or more phosphonocarboxylate and/or phosphinocarboxylate nucleotides (PACE nucleotides).
  • PACE nucleotides are deoxyribonucleotides and the phosphinocarboxylate nucleotides are phosphino acetate nucleotides.
  • the siRNA compound comprises one or more locked nucleic acids (LNA) also defined as bridged nucleic acids or bicyclic nucleotides.
  • LNA locked nucleic acids
  • Preferred locked nucleic acids are 2'-0, 4'-C-ethylene nucleosides (ENA) or 2'-0, 4'-C-methylene nucleosides.
  • LNA and ENA nucleotides are disclosed in WO 98/39352, WO 00/47599 and WO 99/14226, all incorporated herein by reference.
  • the compound comprises one or more altritol monomers (nucleotides), also defined as 1,5 anhydro-2-deoxy-D-altrito- hexitol (see for example, Allart, et al., 1998. Nucleosides & Nucleotides 17: 1523-1526; Herdewijn et al., 1999. Nucleosides & Nucleotides 18: 1371-1376; Fisher et al., 2007, NAR 35(4): 1064- 1074; all incorporated herein by reference).
  • altritol monomers also defined as 1,5 anhydro-2-deoxy-D-altrito- hexitol
  • the present invention explicitly excludes compounds in which each of N and/or N' is a deoxyribonucleotide (d-A, d-C, d-G, d-T).
  • (N)x and (N')y may comprise independently 1, 2, 3, 4, 5, 6, 7, 8, 9 or more deoxyribonucleotides.
  • the present invention provides a compound wherein each of N is an unmodified ribonucleotide and the 3' terminal nucleotide or 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive nucleotides at the 3' terminus of (N')y are deoxyribonucleotides.
  • each of N is an unmodified ribonucleotide and the 5' terminal nucleotide or 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 consecutive nucleotides at the 5' terminus of (N')y are deoxyribonucleotides.
  • the 5' terminal nucleotide or 2, 3, 4, 5, 6, 7, 8, or 9 consecutive nucleotides at the 5' terminus and 1, 2, 3, 4, 5, or 6 consecutive nucleotides at the 3' termini of (N)x are deoxyribonucleotides and each of N' is an unmodified ribonucleotide.
  • (N)x comprises unmodified ribonucleotides and 1 or 2, 3 or 4 consecutive deoxyribonucleotides independently at each of the 5' and 3' termini and 1 or 2, 3, 4, 5 or 6 consecutive deoxyribonucleotides in internal positions; and each of N' is an unmodified ribonucleotide.
  • the 3' terminal nucleotide or 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 13 or 14 consecutive nucleotides at the 3' terminus of (N')y and the terminal 5' nucleotide or 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 13 or 14 consecutive nucleotides at the 5' terminus of (N)x are deoxyribonucleotides.
  • the present invention excludes compounds in which each of N and/or N' is a deoxyribonucleotide.
  • the 5' terminal nucleotide of N or 2 or 3 consecutive of N and 1,2, or 3 of N' is a deoxyribonucleotide.
  • the covalent bond between each consecutive N and N' is a phosphodiester bond.
  • a covalent bond refers to an internucleotide linkage linking one nucleotide monomer to an adjacent nucleotide monomer.
  • a covalent bond includes for example, a phosphodiester bond, a phosphorothioate bond, a P-alkoxy bond, a P-carboxy bond and the like.
  • the normal internucleoside linkage of RNA and DNA is a 3' to 5' phosphodiester linkage.
  • a covalent bond is a phosphodiester bond.
  • Covalent bond encompasses non-phosphorous-containing internucleoside linkages, such as those disclosed in WO 2004/041924 inter alia. Unless otherwise indicated, in preferred embodiments of the structures discussed herein the covalent bond between each consecutive N and N' is a phosphodiester bond.
  • the oligonucleotide sequence of (N)x is fully complementary to the oligonucleotide sequence of (N')y. In other embodiments (N)x and (N')y are substantially complementary. In certain embodiments (N)x is fully complementary to 18-40 consecutive nucleotides in a target mRNA. In other embodiments (N)x is substantially complementary to 18-40 consecutive nucleotides in a target mRNA.
  • neither (N)x nor (N')y are phosphorylated at the 3' and 5' termini. In other embodiments either or both (N)x and (N')y are phosphorylated at the 3' termini (3' Pi). In yet another embodiment, either or both (N)x and (N')y are phosphorylated at the 3' termini with non-cleavable phosphate groups. In yet another embodiment, either or both (N)x and (N')y are phosphorylated at the terminal 2' termini position using cleavable or non-cleavable phosphate groups.
  • the inhibitory nucleic acid molecules of the present invention may comprise one or more gaps and/or one or more nicks and/or one or more mismatches.
  • gaps, nicks and mismatches have the advantage of partially destabilizing the nucleic acid / siRNA, so that it may be more easily processed by endogenous cellular machinery such as DICER, DROSHA or RISC into its inhibitory components.
  • a gap in a nucleic acid refers to the absence of one or more internal nucleotides in one strand
  • a nick in a nucleic acid refers to the absence of an internucleotide linkage between two adjacent nucleotides in one strand.
  • Any of the molecules of the present invention may contain one or more gaps and/or one or more nicks.
  • the present invention provides an otic pharmaceutical composition
  • a therapeutically effective amount of an oligonucleotide compound comprising consecutive nucleotides wherein a first segment of such nucleotides encode a first inhibitory RNA molecule, a second segment of such nucleotides encode a second inhibitory RNA molecule, and a third segment of such nucleotides encode a third inhibitory RNA molecule.
  • Each of the first, the second and the third segment may comprise one strand of a double stranded RNA and the first, second and third segments may be joined together by a linker.
  • the oligonucleotide may comprise three double stranded segments joined together by one or more linker.
  • the present invention provides an otic pharmaceutical composition
  • a therapeutically effective amount of an oligonucleotide comprising consecutive nucleotides which encode three inhibitory RNA molecules; said oligonucleotide may possess a triple stranded structure, such that three double stranded arms are linked together by one or more linker, such as any of the linkers presented hereinabove.
  • This molecule forms a "star"-like structure, and may also be referred to herein as RNAstar.
  • Such structures are disclosed in PCT patent publication WO 2007/091269, assigned to the assignee of the present invention and incorporated herein in its entirety by reference.
  • Said triple- stranded oligonucleotide may be an oligoribonucleotide having the general structure:
  • the above triple stranded structure may have a gap instead of a linker in one or more of the strands.
  • a gap instead of a linker in one or more of the strands.
  • Such a molecule with one gap is technically quadruple stranded and not triple stranded; inserting additional gaps or nicks will lead to the molecule having additional strands.
  • said gapped molecules are more active in inhibiting the target gene than the similar but non-gapped molecules.
  • the present invention provides an otic pharmaceutical composition
  • either or both antisense and sense strands are phosphorylated at the 3' termini.
  • either or both antisense and sense strands are phosphorylated at the 3' termini using non- cleavable phosphate groups.
  • either or both antisense and sense strands are phosphorylated at the terminal 5' termini position using cleavable or non- cleavable phosphate groups. In yet another embodiment, either or both antisense and sense strands are phosphorylated at the terminal 2' termini position using cleavable or non- cleavable phosphate groups.
  • the present invention provides an otic pharmaceutical composition comprising a therapeutically effective amount of an double stranded RNA compound, wherein said double stranded RNA compound is blunt ended and is non-phosphorylated at the termini; however, comparative experiments have shown that double stranded RNA compounds phosphorylated at one or both of the 3'-termini have similar activity in vivo compared to the non-phosphorylated compounds.
  • the otic pharmaceutical compositions of the invention can be prepare using any double stranded RNA sequence disclosed herein and having any of the modifications / structures disclosed herein and any of such compositions can be used in the treatment of the conditions disclosed herein. Unless otherwise indicated, in preferred embodiments of the structures discussed herein the covalent bond between each consecutive N and N' is a phosphodiester bond.
  • an otic pharmaceutical composition of the invention comprises (a) a therapeutically effective amount of at least one oligonucleotide compound which inhibits the expression of a human target gene associated with a disease, a disorder or an injury of the CNS; (b) a permeability enhancer and (c) a pharmaceutically acceptable excipient or carrier, or mixtures thereof.
  • the oligonucleotide sequence of antisense strand is fully complementary to the oligonucleotide sequence of sense. In other embodiments the antisense and sense strands are substantially complementary.
  • the antisense strand is fully complementary to about 18 to about 40 consecutive ribonucleotides a target mRNA set forth in any one of SEQ ID NOS: 1-293. In other embodiments the antisense strand is substantially complementary to about 18 to about 40 consecutive ribonucleotides a target mRNA set forth in any one of SEQ ID NOS: 1-293. In some embodiments the sequence of the antisense strand is substantially complementary to from about 18 to about 40 consecutive ribonucleotides in an mRNA of a target gene associated with a disease, a disorder or an injury of the CNS.
  • the present invention provides an otic pharmaceutical composition comprising an expression vector comprising an antisense oligonucleotide substantially complementary to from about 18 to about 40 consecutive ribonucleotides in an mRNA of a target gene associated with a disease, a disorder or an injury of the CNS.
  • the expression vector further comprises a sense oligonucleotide having complementarity to the antisense oligonucleotide.
  • the present invention further provides a cell comprising an expression vector comprising an antisense oligonucleotide substantially complementary to from about 18 to about 40 consecutive ribonucleotides in an mRNA of a target gene disclosed in Table A.
  • the present invention further provides a siRNA expressed in a cell comprising an expression vector comprising an antisense oligonucleotide disclosed in Table A, a pharmaceutical composition comprising same and use thereof for treatment of any one of the diseases and disorders disclosed herein.
  • the present invention provides an otic pharmaceutical composition
  • a first expression vector comprising an antisense oligonucleotide substantially complementary to from about 18 to about 40 consecutive ribonucleotides in an rnRNA of a target gene disclosed in Table A and a second expression vector comprising a sense oligonucleotide having complementarity to the antisense oligonucleotide comprised in the first expression vector.
  • the present invention further provides a cell comprising a first expression vector comprising an antisense oligonucleotide substantially complementary to from about 18 to about 40 consecutive ribonucleotides in an rnRNA of a target gene disclosed in Table A and a second expression vector comprising a sense oligonucleotide having complementarity to the antisense oligonucleotide comprised in the first expression vector.
  • the present invention further provides a siRNA expressed in a cell comprising such first and second expression vector, a pharmaceutical composition comprising same and use thereof for treatment of any one of the diseases and disorders disclosed herein.
  • Double stranded RNA molecules are prepared essentially as described herein.
  • RNA compounds useful in preparation of the otic pharmaceutical compositions of present invention are synthesized by any of the methods that are well known in the art for synthesis of ribonucleic (or deoxyribonucleic) oligonucleotides. Such synthesis is, among others, described in Beaucage and Iyer, Tetrahedron 1992; 48:2223-2311 ; Beaucage and Iyer, Tetrahedron 1993; 49: 6123-6194 and Caruthers, et. al., Methods Enzymol. 1987; 154: 287-313; the synthesis of thioates is, among others, described in Eckstein, Ann. Rev. Biochem.
  • oligonucleotides useful in preparation of the otic pharmaceutical compositions of the present invention can be synthesized separately and joined together post- synthetically, for example, by ligation (Moore et al., 1992, Science 256, 9923; Draper et al., International Patent Publication No. WO 93/23569; Shabarova et al., 1991, NAR 19, 4247; Bellon et al., 1997, Nucleosides & Nucleotides, 16, 951; Bellon et al., 1997, Bioconjugate Chem. 8, 204), or by hybridization following synthesis and/or deprotection.
  • oligonucleotides are prepared according to the sequences disclosed herein. Overlapping pairs of chemically synthesized fragments can be ligated using methods well known in the art (e.g., see US Patent No. 6,121,426). The strands are synthesized separately and then are annealed to each other in the tube. Then, the double- stranded siRNAs are separated from the single- stranded oligonucleotides that were not annealed (e.g. because of the excess of one of them) by HPLC. In relation to the siRNAs or siRNA fragments of the present invention, two or more such sequences can be synthesized and linked together for use in the present invention.
  • the double stranded RNA compounds useful in preparation of the otic pharmaceutical compositions of the invention can also be synthesized via tandem synthesis methodology, as described for example in US Patent Publication No. US 2004/0019001, wherein both siRNA strands are synthesized as a single contiguous oligonucleotide fragment or strand separated by a cleavable linker which is subsequently cleaved to provide separate siRNA fragments or strands that hybridize and permit purification of the siRNA duplex.
  • the linker is selected from a polynucleotide linker or a non-nucleotide linker.
  • the oligonucleotide compounds of the present invention While it is possible for the oligonucleotide compounds of the present invention to be administered into the ear as the raw chemical, it is preferable to present them as an otic pharmaceutical composition.
  • the one or more oligoribonucleotide compounds are produced by endogenous intracellular complexes.
  • the instant invention relates to a method of treatment of a disease, a disorder or an injury of the CNS in a subject comprising administering to the ear canal of the subject a therapeutically effective amount of an otic pharmaceutical composition comprising one or more therapeutic double stranded RNA compounds.
  • the method of the invention recognizes that otic administration of a therapeutically effective amount of one or more double stranded RNA compounds according to the invention is useful to treat or prevent a disease, a disorder or an injury of the CNS in a subject in need thereof.
  • the present invention provides an otic pharmaceutical composition comprising one or more inhibitory oligonucleotide compounds in an amount effective to down-regulate expression of a mammalian or non-mammalian gene in the CNS of a subject suffering from a disease, a disorder or an injury of the CNS and a pharmaceutically acceptable carrier.
  • the mammalian gene is a human gene.
  • the non- mammalian gene is involved in a mammalian disease, preferably human disease.
  • the subject is a human.
  • the invention further provides an otic pharmaceutical composition comprising at least one double stranded RNA compound which reduces or inhibits expression of a gene selected from genes disclosed in Table A; and a pharmaceutically acceptable carrier.
  • the double stranded RNA compounds are processed intracellularly by endogenous cellular complexes to produce one or more oligoribonucleotides compounds.
  • the invention further provides an otic pharmaceutical composition comprising one or more of chemically modified double stranded RNA compounds in an amount effective to inhibit expression in a CNS cell of a target gene that is associated with a disease, a disorder or an injury of the CNS, the double stranded RNA compound comprising a sequence which is substantially complementary to the sequence of the mRNA of the target gene, and a pharmaceutically acceptable carrier.
  • the invention further provides an otic pharmaceutical composition comprising one or more inhibitory oligonucleotide compounds; a permeability enhancer and a pharmaceutically acceptable vehicle or carrier.
  • the composition comprises a mixture of two or more different oligonucleotides / siRNA compounds.
  • the penetration enhancer is selected from any compound or any combination of two ore more compounds that enhance the penetration of a therapeutic oligonucleotide through the skin and/or the tympanic membrane in the ear of a subject suffering from or at risk of a disease, a disorder or an injury of the CNS.
  • the penetration / permeability enhancer is selected from, without being limited to, polyethylene glycol (PEG), glycerol (glycerin), maltitol, sorbitol etc.; diethylene glycol monoethyl ether, azone, benzalkonium chloride (ADBAC), cetylperidium chloride, cetylmethylammonium bromide, dextran sulfate, lauric acid, menthol, methoxysalicylate, oleic acid, phosphatidylcholine, polyoxyethylene, polysorbate 80, sodium glycholate, sodium lauryl sulfate, sodium salicylate, sodium taurocholate, sodium taurodeoxycholate, sulfoxides, sodium deoxycholate, sodium glycodeoxycholate, sodium taurocholate and surfactants such as sodium lauryl sulfate, laureth-9, cetylpyridinium chloride and polyoxyethylene monoalkyl ethers, benzo
  • the permeability enhancer is a polyol.
  • the oligonucleotide is in admixture with a polyol.
  • Suitable polyols for inclusion in the solutions of the invention include glycerol and sugar alcohols such as sorbitol, mannitol or xylitol, polyethylene glycol and derivatives thereof.
  • the otic pharmaceutical compositions of the present invention also include one or more of various other pharmaceutically acceptable ingredients, such as, without being limited to, one ore more of buffering agent, preservative, surfactant, carrier, solvent, diluent, co-solvent, viscosity building/enhancing agent, excipient, adjuvant and vehicle.
  • buffering agent preservative
  • surfactant such as benzalkonium chloride and disodium edetate (EDTA) are included in the compositions of the invention in concentrations sufficient for effective antimicrobial action, about 0.0001 to 0.1%, based on the weight of the composition.
  • EDTA disodium edetate
  • the polyol is glycerol.
  • glycerol is present at a final concentration of about 0.1% to about 35%; about 1% to about 30%; about 5% to about 25%, preferably about 10% to about 20% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 2%, 2.5%, 5%, 10%, 12.5%, 15%, 17.5%, 20%, 22.5%, 25%, 27.5% or about 30% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 2% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 10% by volume of the otic pharmaceutical composition.
  • the final concentration of glycerol in the pharmaceutical composition is about 20% by volume of the otic pharmaceutical composition.
  • the pharmaceutical composition is brought to about the subject's body temperature, which is about 30°C to about 38°C, prior to application to the ear.
  • the otic oligonucleotide composition are formulated for topical administration to the external ear, including the ear canal of a subject by any suitable mode of administration.
  • suitable modes of administration of the otic pharmaceutical compositions of the invention include invasive and non-invasive modes of administration, such as without being limited to, instillation (of ear drop solution), injection (of injectable formulation), deposition (of solid or semi-solid formulation, e.g. ointment, gel), infusion or spraying into the ear.
  • the compositions of the invention are sprayed into the ear canal, producing a semi- solid (i.e. a foam or mousse) in the ear canal of the subject.
  • compositions of the invention can also be impregnated in a porous media (for example, an ear wick such as a sponge, gauze, cotton, or hydrocellulose), which is suitable for insertion into the external ear canal to the tympanic membrane.
  • a porous media for example, an ear wick such as a sponge, gauze, cotton, or hydrocellulose
  • the compositions of the present invention are administered topically into the ear canal as ear drops or injected through a cannula into the ear canal or sprayed into the ear canal or injected through the tympanic membrane (transtympanic injection).
  • the pharmaceutical composition is applied to the ear canal when the subject's head is tilted to one side and the treated ear is facing upward.
  • the subject remains with his head tilted to one side with the treated ear facing upwards for a sufficient time to allow penetration of the otic pharmaceutical composition.
  • the pharmaceutical composition is applied to the ear using a receptacle for eardrops, for example using a dropper of for example, 10-100 microliter per drop, or a wick.
  • the compositions of the present invention are brought to about the subject's body temperature, which is about 30°C to about 38°C, prior to application to the ear. Delivery can be effected by any mean (e.g. drops, spray), using any effective instrument for placing the composition inside the ear canal or on/adjacent to the tympanic membrane) or for injecting the composition (e.g. through the tympanic membrane).
  • the at least one double stranded RNA compound is delivered to the ear of a subject in an otic pharmaceutical composition designed for topical non-invasive administration (e.g. instillation, deposition, spraying, insertion, transdermal patch).
  • the otic pharmaceutical composition described herein is any conventional form, such as, without being limited to, a cream, a foam, a paste, an ointment, an emulsion, a solution, a gel, an hydrogel, a spray, a suspension, a microemulsion, microspheres, microcapsules, nanospheres, nanoparticles, lipid vesicles, liposomes, polymeric vesicles, patches, biological inserts, transdermal patch.
  • the present invention provides a non-invasive method of attenuating expression of a target mRNA in a subject suffering from a disease, a disorder or an injury of the CNS, which comprises topically administering into the ear canal of the subject a pharmaceutical composition formulated as an ear drop comprising at least one oligonucleotide compound directed to the target mRNA, in an amount and over a period of time effective to attenuate expression of the target mRNA in the CNS of the subject.
  • the present invention provides a method of treating a disease, a disorder or an injury of the CNS in a subject in need thereof, which comprises topically administering to the ear canal of the subject a pharmaceutical composition formulated as an ear drop, comprising at least one oligonucleotide directed to a target gene associated with the disease, the disorder or the injury of the CNS, in an amount and over a period of time effective to treat the subject.
  • the target mRNA is a mammalian or a non-mammalian mRNA.
  • the mammalian mRNA is a human mRNA.
  • the non- mammalian mRNA is a product of a gene involved in a mammalian disease, preferably human disease.
  • the otic pharmaceutical composition according to the present invention comprises a single type of double stranded RNA compound directed to a target gene associated with the disease, the disorder or the injury of the CNS. In some embodiments the otic pharmaceutical composition according to the present invention comprises two or more different types of double stranded RNA compounds directed to a target gene associated with the disease, the disorder or the injury of the CNS. In some embodiments, simultaneous inhibition of the target gene by two or more different types of double stranded RNA compounds additive or synergistic effect for treatment of the diseases disclosed herein. In other embodiments otic pharmaceutical composition of the invention comprises two or more different types of double stranded RNA compounds directed to two or more target genes associated with the disease, the disorder or the injury of the CNS. In some embodiments, simultaneous inhibition of more than one target gene by the otic pharmaceutical composition of the invention provides additive or synergistic effect for treatment of the diseases disclosed herein.
  • the present invention provides an otic pharmaceutical composition
  • an otic pharmaceutical composition comprising a therapeutically effective amount of an oligonucleotide compound, wherein the oligonucleotide compound is linked or bound (covalently or non-covalently) to an antibody or aptamer against cell surface internalizable molecules expressed on the target cells, in order to achieve enhanced targeting for treatment of the diseases disclosed herein.
  • an aptamer which acts like a ligand/antibody is combined (covalently or non-covalently) with a double stranded RNA compound in preparation of otic pharmaceutical compositions according to the present invention.
  • the present invention also provides for a process for preparing an otic pharmaceutical composition of the invention, in accordance with formulation techniques known to those skilled in the art.
  • the process for preparing an otic pharmaceutical composition of the invention comprises combining, in any suitable order, a therapeutically effective amount of at least one oligonucleotide compound, one or more permeability enhancer and at least one pharmaceutically acceptable excipient or carrier, or mixtures thereof, such a composition preferably having extended chemical and/or physical stability as described herein.
  • the process for preparing an otic pharmaceutical composition of the invention comprises combining, in any suitable order, a therapeutically effective amount of at least one oligonucleotide compound, one or more permeability enhancer, at least one pharmaceutically acceptable excipient or carrier, or mixtures thereof and an antibacterial agent and/or preservative.
  • the otic pharmaceutical composition includes a pharmacologically acceptable surfactant to assist in dissolving the double stranded RNA compound.
  • an otic pharmaceutical composition of the invention further comprises an additional therapeutically active agent, such compositions being useful in combination therapies as described herein.
  • the additional pharmaceutically active agent is selected from, without being limited to, such as non-steroidal anti- inflammatory drugs, corticosteroids, antifungal, antibiotics, and the like.
  • the otic pharmaceutical compositions of the invention comprise a therapeutically effective amount of at least one double stranded RNA compound which inhibits the expression of a target gene associated with a disease, a disorder or an injury of the CNS, or salt thereof, in an amount ranging from about 0.1 mg/ml to about 100 mg/ml of the composition.
  • the amount of at least one double stranded RNA compound ranges from between about 1 mg/ml to about 50 mg/ml of the otic pharmaceutical composition.
  • the amount of at least one double stranded RNA compound ranges from between about 5 mg/ml to about 20 mg/ml of the otic pharmaceutical composition.
  • a pharmaceutically acceptable excipient or carrier is selected from a physiologically acceptable aqueous carrier, such as water, sodium chloride, buffer, saline (e.g. phosphate buffered saline (PBS)), mannitol, and the like, physiologically acceptable nonaqueous carrier, such as oil, and combinations thereof.
  • physiologically acceptable aqueous carrier such as water, sodium chloride, buffer, saline (e.g. phosphate buffered saline (PBS)), mannitol, and the like
  • physiologically acceptable nonaqueous carrier such as oil, and combinations thereof.
  • Suitable aqueous and/or non-aqueous pharmaceutically acceptable carrier or vehicle is one that has no unacceptably injurious or toxic effect on the subject when administered as a component of a composition in an amount required herein. No excipient ingredient of such a carrier or vehicle reacts in a deleterious manner with another excipient or with the therapeutic oligonucleotide compound in a
  • the present invention provides an otic pharmaceutical composition according to the present invention for treating a disease, a disorder or an injury of the CNS.
  • the present application provides for delivery of therapeutic oligonucleotide compounds to the CNS of a subject suffering from a disease, a disorder or injury of the CNS, by direct application of an otic pharmaceutical composition to the outer ear of the subject.
  • the pharmaceutical composition is applied to the ear canal. Delivery to the ear is also refereed to as aural or otic delivery.
  • the otic pharmaceutical compositions according to the invention comprise double stranded RNA compound in liposome or lipofectin formulations and the like.
  • Such formulations can be prepared by methods well known to those skilled in the art. Such methods are described, for example, in US Patent Nos. 5,593,972, 5,589,466, and 5,580,859, which are herein incorporated by reference.
  • compositions in liposomes benefits absorption.
  • the otic pharmaceutical compositions comprise double stranded RNA compound formulated with polyethylenimine (PEI), with PEI derivatives, e.g. oleic and stearic acid modified derivatives of branched PEI, with chitosan or with poly(lactic- co-glycolic acid) (PLGA).
  • PEI polyethylenimine
  • PEI derivatives e.g. oleic and stearic acid modified derivatives of branched PEI
  • PLGA poly(lactic- co-glycolic acid)
  • Formulating the compositions in e.g. liposomes, micro- or nano- spheres and nanoparticles may enhance stability and benefit absorption. Examples of delivery systems useful in the present invention include U.S. Patent Nos.
  • liquid forms are designed for administration as eardrops, also referred to as otic drops or aural drops.
  • liquid compositions include aqueous solutions, with and without organic co-solvents, aqueous or oil suspensions, emulsions with edible oils, as well as similar pharmaceutical vehicles.
  • the otic pharmaceutical compositions are in a from that remains in the ear canal of a subject for at least about 30 min without leakage of the composition out of the canal.
  • the otic pharmaceutical composition of the invention is designed for administration as eardrops and the subject receiving the ear drops keep his head on the side with the treated ear facing upward to prevent leakage of the drop out of the canal.
  • Additional formulations for improved delivery of the compounds of the present invention can include conjugation of double stranded RNA molecules to a targeting molecule.
  • the conjugate is usually formed through a covalent attachment of the targeting molecule to the sense strand of the double stranded RNA, so as not to disrupt silencing activity.
  • Potential targeting molecules useful in the present invention include proteins, peptides and aptamers, as well as natural compounds, such as e.g. cholesterol.
  • conjugation to a protamine fusion protein has been used (see for example: Song et al., Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors, Nat Biotechnol. 2005. 23(6):709-17).
  • compositions of the present invention are administered and dosed in accordance with good medical practice, taking into account the clinical condition of the individual patient, the disease to be treated, the site and method of administration, scheduling of administration, patient age, sex, body weight and other factors known to medical practitioners.
  • a “therapeutically effective dose” or a “therapeutic effective amount” refers to an amount of a pharmaceutical compound or composition which is effective to achieve an improvement in a subject or his physiological systems including, but not limited to, improved survival rate, more rapid recovery, suppressed progress of the disease, or improvement or elimination of symptoms, and other indicators as are selected as appropriate determining measures by those skilled in the art.
  • a “therapeutically effective dose” or a “therapeutic effective amount” for purposes herein is thus determined by such considerations as are known in the art.
  • the dose must be effective to achieve improvement including but not limited to improved survival rate or more rapid recovery, or improvement or elimination of symptoms and other indicators as are selected as appropriate measures by those skilled in the art.
  • the otic pharmaceutical compositions of the invention are administered in a single dose or in multiple doses.
  • Dosage to the ear is determined, inter alia, by the activity of the oligonucleotide, the indication and the severity of the disorder and comprises administering a dose of about 0.1 ng to about 10 mg, about 1 ng to about 1 mg, or about 10 ng to about 1 mg, total oligonucleotide in pharmaceutically acceptable excipient or carrier.
  • the concentration of double stranded RNA compound in the composition is between 0.1 mg/ml to 100 mg/ml, preferably between 1 mg/ml to 100 mg/ml, and more preferably between 5 mg/ml to 20 mg/ml.
  • the active dose of oligonucleotide compound for humans is in the range of from lng/kg to about 20-100 mg/kg body weight per day, preferably about 0.01 mg to about 2-10 mg/kg body weight per day, in a regimen of a single dose or multiple doses administered in one dose per day or twice or three or more times per day for a period of 1-4 weeks or longer, or even for the life of the subject.
  • the otic pharmaceutical compositions of the present invention are administered to the external ear, including the ear canal of a subject by any suitable mode of administration.
  • Suitable modes of administration of the otic oligonucleotide compositions of the invention include invasive and non-invasive mode of administration, such as without being limited to, instillation (of ear drops), injection, deposition, or spraying into the ear.
  • the compositions of the present invention are administered topically into the ear canal as ear drops or injected through a cannula into the ear canal or injected through the tympanic membrane (transtympanic injection).
  • compositions of the present invention are warmed to a temperature of about 30° C to about 38°C prior to administration into the ear of the subject.
  • the mode of administration may depend on many factors, including without being limited to, the affected CNS regions, nature and severity of the CNS disease or condition or injury being treated, as well as other clinical conditions of the individual subject.
  • Otic (inner ear or cochlear) implants comprising the double stranded RNA compounds are also useful.
  • the otic pharmaceutical compositions of the invention are delivered to the CNS in an amount effective to provide a protective or therapeutic effect.
  • protective or therapeutic effects include inhibition of target protein expression or knockdown of at least one target gene.
  • inhibiting expression of at least one target gene confers upon the cells and/or tissues of the CNS neuroprotective properties.
  • the otic pharmaceutical compositions of the invention are administered in any form that allows the active ingredient(s) (i.e. at least one oligonucleotide compound) to prevent, suppress, ameliorate, or otherwise treat the CNS diseases and conditions disclosed herein.
  • active ingredient(s) i.e. at least one oligonucleotide compound
  • the otic pharmaceutical compositions can be formulated as a cream, foam, paste, ointment, emulsion, liquid solution, gel, spray, suspension, microemulsion, microspheres, microcapsules, nanospheres, nanoparticles, lipid vesicles, liposomes, polymeric vesicles, patches, biological inserts, aerosol, polymeric or polymeric-like material and/or any other form known in the art, including any form suitable for known or novel pharmaceutical delivery systems or devices, such as a removable and/or absorbable, dissolvable, and/or degradable implant.
  • Sterile liquid pharmaceutical compositions, solutions or suspensions can be utilized invasively, for example, by transtympanic injection; or topically, e.g. by ear drop, foam, spray, gel, cream, ointment, application into the ear canal.
  • the liquid compositions include aqueous solutions, with and without organic co-solvents, aqueous or oil suspensions, emulsions e.g. with edible oils, as well as similar pharmaceutical vehicles.
  • the otic compositions of the invention circumvent the blood-brain barrier (BBB) and are delivered directly to the CNS.
  • BBB blood-brain barrier
  • the otic pharmaceutical composition of the invention is useful for delivery of the double stranded RNA compound directly into the CNS by transport along a neural pathway to the CNS, or by way of a perivascular channel, a prelymphatic channel, or a lymphatic channel associated with the brain, retina, optic nerve and/or spinal cord.
  • the otic pharmaceutical composition of the invention delivers the double stranded RNA compound to the cerebrospinal fluid and then subsequently to the CNS, including the brain, retina, optic nerve and/or spinal cord.
  • the otic pharmaceutical compositions of the present invention comprising one or more chemically modified double stranded RNA compounds are delivered to the CNS by direct application of the pharmaceutical composition to the outer ear. Delivery to the ear may also be refereed to as aural or otic delivery. In various embodiments the otic pharmaceutical composition is applied to the ear canal. In some embodiments the otic pharmaceutical compositions of the invention are formulated as sterile liquid pharmaceutical compositions, solutions or suspensions and are utilized invasively, for example, by transtympanic injection.
  • the otic pharmaceutical compositions of the invention are formulated for topical non-invasive administration, as cream, foam, paste, ointment, emulsion, solution, gel, spray, suspension, microemulsion, microspheres, microcapsules, nanospheres, nanoparticles, lipid vesicles, liposomes, polymeric vesicles, patches, biological inserts, for instillation, deposition or spraying into the ear canal.
  • the otic pharmaceutical compositions of the invention are formulated for administration as ear drops (also referred to as otic drops or aural drops).
  • the eardrops remain in the ear canal of the subject for about 30 minutes. It is thus preferable that the subject receiving the drops keeps his head on the side to prevent leakage of the drop out of the canal.
  • the present invention provides a method of treating a subject afflicted with a disease, a disorder or an injury of the CNS, which comprises administering to the ear of the subject an otic composition comprising at least one therapeutic agent in an amount and over a period of time effective to treat the subject.
  • the therapeutic agent is an oligonucleotide compound, including chemically synthesized siRNA.
  • the present invention provides a method of treating a disease, a disorder or an injury of the CNS in a subject in need thereof, which comprises administering to the ear of the subject an otic pharmaceutical composition comprising at least one oligonucleotide compound directed to a target gene associated with the disease, the disorder or the injury of the CNS, in an amount and over a period of time effective to treat the subject.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from a disease, a disorder or an injury of the CNS, which comprises administering to the ear of the subject an otic pharmaceutical composition comprising at least one oligonucleotide directed to the target mRNA, in an amount and over a period of time effective to attenuate expression of the target mRNA in the CNS of the subject.
  • the present invention provides a non-invasive method of delivery of a therapeutic oligonucleotide to a CNS in a subject suffering from a disease, a disorder or an injury of the CNS comprising topically and non-invasively administering an otic oligonucleotide composition to the ear of the subject.
  • the present invention provides a method of effecting neuroprotection to cells of the CNS in a subject in need thereof comprising administering to the subject's ear canal a therapeutic agent and an otic carrier in an amount and over a period of time effective to effect neuroprotection in the subject.
  • the cells are retinal cells, in particular retinal ganglion cells.
  • the cells are optic nerve cells.
  • the cells are spinal cord cells or brain cells.
  • the disease disorder or injury of the CNS is associated with APP, MAPT, SOD1, BACE1, CASP3, TGM2, TARDBP, ADRB1, CAMK2A, CBLN1, CDK5R1, GABRA1, MAPK10, NOS1, NPTX2, NRGN, NTS, PDCD2, PDE4D, PENK, SYT1, TTR, FUS, LRDD, CYBA, ATF3, CASP2, HRK, C1QBP, BNIP3, MAPK8, MAPK14, Racl, GSK3B, P2RX7, TRPM2, PARG, CD38, STEAP4, BMP2, GJA1, TYROBP, CTGF, ANXA2, DUOX1, RTP801, RTP801L, NOX4, NOX1, NOX2 (gp91pho, CYBB), NOX5, DUOX2, NOXOl, NOX02 (p47phox, NCF1), NOXA1, NOXA2 (p67phox, NCF
  • the present invention relates to a method for the treatment of a subject in need of treatment for a disease or disorder or injury or symptom or condition associated with the disease or disorder, associated with the expression of a gene selected from the group consisting of APP, MAPT, SOD1, BACE1, CASP3, TGM2, TARDBP, ADRB1, CAMK2A, CBLN1, CDK5R1, GABRA1, MAPK10, NOS1, NPTX2, NRGN, NTS, PDCD2, PDE4D, PENK, SYT1, TTR, FUS, LRDD, CYBA, ATF3, CASP2, HRK, C1QBP, BNIP3, MAPK8, MAPK14, Racl, GSK3B, P2RX7, TRPM2, PARG, CD38, STEAP4, BMP2, GJA1, TYROBP, CTGF, ANXA2, DUOX1, RTP801, RTP801L, NOX4, NOX1, NOX2 (gp91pho, CY
  • the therapeutic oligonucleotide is a siRNA compound, preferably chemically modified according to the embodiments of the present invention.
  • the subject being treated is a warm-blooded animal and, in particular a mammal, and preferably a human.
  • Treating a subject refers to administering to the subject a therapeutic substance effective to alleviate symptoms associated with a disease or condition, to delay the onset of the disease, to slow the progress of the disease, to lessen the severity or cure the disease, or to prevent the disease from occurring.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent a disorder, to slow the progress of a disease or to reduce the symptoms of a disorder.
  • compositions of the invention are administered before, during or subsequent to the onset of the disease or condition.
  • the invention provides a method of down-regulating the expression of a target gene by at least 30% as compared to a control, comprising contacting a target gene mRNA with one or more of the chemically modified double stranded RNA compound of the otic pharmaceutical compositions of the present invention.
  • topical administration of an otic pharmaceutical composition of the invention which comprises at least one oligonucleotide directed to a target gene associated with the disease, the disorder or the injury of the CNS, inhibits or down-regulates the mammalian APP, MAPT, SOD1, BACE1, CASP3, TGM2, TARDBP, ADRB1, CAMK2A, CBLN1, CDK5R1, GABRA1, MAPK10, NOS1, NPTX2, NRGN, NTS, PDCD2, PDE4D, PENK, SYT1, TTR, FUS, LRDD, CYBA, ATF3, CASP2, HRK, C1QBP, BNIP3, MAPK8, MAPK14, Racl, GSK3B, P2RX7, TRPM2, PARG, CD38, STEAP4, BMP2, GJA1, TYROBP, CTGF, ANXA2, DUOX1, RTP801, RTP801L, NOX4, NOX1, NOX2 (gp91
  • the present invention provides a method of inhibiting the expression of a target gene in a subject suffering from a disease, a disorder or an injury of the CNS, which comprises administering to the ear of the subject an otic pharmaceutical composition comprising at least one oligonucleotide directed to the target gene, in an amount and over a period of time effective to inhibit expression of the target gene in the CNS of the subject.
  • the invention provides a method of inhibiting the expression of a target gene in a subject suffering from a disease, a disorder or an injury of the CNS, by at least 30%, preferably by 40%, 50%, 60% or 70%, more preferably by 75%, 80% or 90% as compared to a control, which comprises administering to the ear of the subject an otic pharmaceutical composition comprising at least one oligonucleotide directed to the target gene, in an amount and over a period of time effective to inhibit expression of the target gene in the CNS of the subject.
  • the invention provides a method of inhibiting the expression of the target gene by at least 30%, preferably by 40%, 50%, 60% or 70%, more preferably by 75%, 80% or 90% as compared to a control comprising contacting an mRNA transcript of the target gene with one or more of the double stranded RNA compounds of the otic pharmaceutical compositions of the invention.
  • the effect of inhibition of a target gene by the otic pharmaceutical compositions comprising an oligonucleotide inhibitor is determined by examining siRNA effect on the mRNA or on the corresponding protein product of the target gene, whereby the inhibition is selected from the group comprising inhibition of function (which is examined by, for example, an enzymatic assay or a binding assay with a known interactor of the native gene / polypeptide, inter alia), inhibition of protein (which is examined by, for example, Western blotting, ELISA or immuno- precipitation, inter alia) and inhibition of target gene mRNA expression (which is examined by, for example, Northern blotting, quantitative RT-PCR, in-situ hybridization or microarray hybridization, inter alia).
  • an oligonucleotide inhibitor e.g. chemically modified siRNA compound
  • an otic pharmaceutical composition of the invention comprising at least one chemically modified double stranded RNA compound is down-regulating the target gene or polypeptide, whereby the down-regulation is selected from the group comprising down- regulation of function (which is examined by, for example, an enzymatic assay or a binding assay with a known interactor of the native gene / polypeptide, inter alia), down-regulation of protein (which is examined by, for example, Western blotting, ELISA or immuno- precipitation, inter alia) and down-regulation of target gene mRNA expression (which is examined by, for example, Northern blotting, quantitative RT-PCR, in-situ hybridization or microarray hybridization, inter alia).
  • down-regulation of function which is examined by, for example, an enzymatic assay or a binding assay with a known interactor of the native gene / polypeptide, inter alia
  • down-regulation of protein which is examined by, for example, Western blotting, ELISA or immuno- precipitation, inter
  • the invention provides a method of treating a subject suffering from or susceptible to any disease or disorder or injury of the CNS accompanied by an elevated level of a mammalian target gene associated with a disease, a disorder or an injury of the CNS, which comprises topically administering to the canal of the subject's ear an otic pharmaceutical composition comprising: (a) a therapeutically effective amount of at least one oligonucleotide compound which inhibits the expression of a mammalian target gene associated with a disease, a disorder or an injury of the CNS; (b) a permeability enhancer or mixtures thereof and (c) a pharmaceutically acceptable excipient or carrier, or mixtures thereof, thereby treating the subject.
  • the mammalian target gene is a human target gene.
  • the target gene is selected from one or more of SEQ ID NO: 1-293.
  • otic pharmaceutical compositions comprising chemically modified double stranded RNA compounds which inhibit a mammalian target gene associated with a disease, a disorder or an injury of the CNS, or polypeptide expression thereof, are discussed herein at length, and any of said double stranded RNA molecules and/or pharmaceutical compositions are beneficially employed in the treatment of a subject suffering from or susceptible to any of said conditions.
  • the method of the invention further includes non-invasive administration of an otic pharmaceutical composition comprising a therapeutically effective amount of one or more of chemically modified double stranded RNA compounds which down-regulate expression of a target gene associated with a disease, a disorder or an injury of the CNS.
  • the target gene is selected from the group consisting of the genes set forth in Table A.
  • the methods of treating the diseases disclosed herein include administering a novel otic pharmaceutical composition comprising at least one chemically modified double stranded RNA compound directed to a target gene associated with a disease, a disorder or an injury of the CNS in conjunction or in combination with an additional inhibitor directed to a target gene associated with a disease, a disorder or an injury of the CNS, and/or a substance which improves the pharmacological properties of the chemically modified double stranded RNA compound, and/or an additional compound known to be effective in the treatment of a subject suffering from or susceptible to neurodegenerative disease, a neurological disorder, a malignancy or a tumor, an affective disorder, or nerve damage resulting from a cerebrovascular disorder, injury, or infection of the CNS.
  • Combination therapies comprising known treatments for treating a subject suffering from or affected by or susceptible to diseases, disorders or injury of the CNS, in conjunction with the novel otic pharmaceutical compositions and therapies described herein are considered part of the current invention.
  • the otic pharmaceutical compositions of the invention further comprise a known therapeutically active compound which is directed to treatment of CNS conditions (e.g. Cholinesterase inhibitors, glutamate regulators, etc.).
  • a known therapeutically active compound which is directed to treatment of CNS conditions (e.g. Cholinesterase inhibitors, glutamate regulators, etc.).
  • Appropriate therapeutic amount of such a known second therapeutic agents for use in combination with an otic pharmaceutical composition of the invention are readily appreciated by those skilled in the art.
  • an otic pharmaceutical composition of the invention is carried out by any of the many known routes of administration, including invasive and noninvasive methods of administration, as determined by a skilled practitioner.
  • routes of administration including invasive and noninvasive methods of administration, as determined by a skilled practitioner.
  • Using specialized formulations it is possible to administer the compositions, inter alia, by instillation (e.g. of ear drops), injection, deposition, or spraying into the ear.
  • a second therapeutic agent is administered by any suitable route, for example, by otic, oral, buccal, inhalation, sublingual, rectal, vaginal, transurethral, nasal, topical, percutaneous (i.e., transdermal), or parenteral (including intravenous, intramuscular, subcutaneous, and intracoronary) administration.
  • an oligonucleotide of the invention and the second therapeutic agent/composition are administered by the same route, either provided in a single composition or as two or more different pharmaceutical compositions.
  • a different route of administration for the novel otic pharmaceutical compositions of the invention and the second therapeutic composition/agent is either possible or preferred. Persons skilled in the art are aware of the best modes of administration for each therapeutic agent, either alone or in combination.
  • the present invention provides an otic pharmaceutical composition comprising two or more double stranded RNA molecules for the treatment of any of the diseases and conditions mentioned herein.
  • the two or more double stranded RNA molecules or formulations comprising said molecules are admixed in the pharmaceutical composition in amounts which generate equal or otherwise beneficial activity.
  • the two or more double stranded RNA molecules are covalently or non-covalently bound, or joined together by a nucleic acid linker of a length ranging from 2- 100, preferably 2-50 or 2-30 nucleotides.
  • the two or more double stranded RNA molecules target mRNA to APP, MAPT, SOD1, BACE1, CASP3, TGM2, TARDBP, ADRB1, CAMK2A, CBLN1, CDK5R1, GABRA1, MAPK10, NOS1, NPTX2, NRGN, NTS, PDCD2, PDE4D, PENK, SYT1, TTR, FUS, LRDD, CYBA, ATF3, CASP2, HRK, C1QBP, BNIP3, MAPK8, MAPK14, Racl, GSK3B, P2RX7, TRPM2, PARG, CD38, STEAP4, BMP2, GJA1, TYROBP, CTGF, ANXA2, DUOX1, RTP801, RTP801L, NOX4, NOX1, NOX2 (gp91pho, CYBB), NOX5, DUOX2, NOXOl, NOX02 (p47phox, NCF1), NOXA1, NOXA2 (p
  • At least one of the two or more double stranded RNA compounds target the genes set forth in Table A.
  • the pharmaceutical compositions of the invention further comprise one or more additional double stranded RNA molecule, which targets one or more additional target gene.
  • simultaneous inhibition of said additional gene(s) provides an additive or synergistic effect for treatment of the diseases disclosed herein.
  • the treatment regimen according to the invention is carried out, in terms of administration mode, timing of the administration, and dosage, so as to thereby treat a subject suffering from or susceptible to neurodegenerative disease, a neurological disorder, a malignancy or a tumor of the CNS, an affective disorder, or nerve damage resulting from a cerebrovascular disorder, injury, or infection of the CNS.
  • the otic pharmaceutical compositions of the invention are useful in treating or preventing various diseases, disorders and injury that affect the central nervous system (CNS), such as, without being limited to, the diseases, disorders and injury that are disclosed herein below.
  • CNS central nervous system
  • oligonucleotide compounds Delivery of therapeutic oligonucleotide compounds via the ear is useful in the treatment of a broad spectrum of eye diseases and disorders in which neuroprotection of the optic nerve would be of benefit, for example in: open angle primary/secondary glaucoma, multiple sclerosis (optic neuritis), central or brunch retinal vein occlusion, ischemic optic neuropathy ( in status epilepticus, HIV-1 infection), optic nerve injury, tumors extending into the suprasellar region (above the sella turcica), juxta chiasmal tumors (the visual loss associated with compression of the optic chiasm by pituitary tumors may be transient or permanent, possibly related to the extent of irreversible retrograde degeneration to the retinal ganglion cells, Retinoblastoma.
  • open angle primary/secondary glaucoma multiple sclerosis (optic neuritis), central or brunch retinal vein occlusion, ischemic optic neuropathy ( in status epilepticus
  • the eye disorder, disease or injury is selected from glaucoma, diabetic retinopathy (DR), diabetic macular edema (DME), age related macular degeneration (AMD) Leber's hereditary optic neuropathy (LHON) or Leber optic atrophy.
  • the disorder is a primary glaucoma, selected from primary open angle glaucoma, normal- tension glaucoma or angle-closure glaucoma.
  • the disorder is a progressing glaucoma.
  • the disorder is a secondary glaucoma selected from pseudoexfoliation glaucoma, pigmentary glaucoma, neovascular glaucoma, steroid- induced glaucoma, acute angle closure glaucoma or treatment refractory glaucoma.
  • the ocular disorder, disease or injury is optic neuritis, central retinal vein occlusion, brunch retinal vein occlusion (BRVO).
  • the eye disorder, disease or injury is retinitis pigmentosa (RP), ischemic optic neuropathy or optic nerve injury.
  • the optic neuropathy is selected from non-arteritic anterior ischemic optic neuropathy (NAION), optic neuritis, neuromyelitis optica, dominant optic atrophy, Leber's hereditary optic neuropathy.
  • NAION non-arteritic anterior ischemic optic neuropathy
  • optic neuritis neuromyelitis optica
  • dominant optic atrophy Leber's hereditary optic neuropathy.
  • ocular disorder, disease or injury is retinopathy of prematurity (ROP) retinal ganglion degeneration, macular degeneration, hereditary optic neuropathy, metabolic optic neuropathy, optic neuropathy due to a toxic agent or neuropathy caused by adverse drug reactions or vitamin deficiency.
  • ROP retinopathy of prematurity
  • the disorder is vision loss associated with a tumor.
  • POAG primary-open-angle glaucoma
  • POAG results from a build up of aqueous humor fluid within the anterior chamber of the eye resulting in intraocular pressure (IOP).
  • Elevated IOP which can be measured by a "tonometry" test, results from fluid entering the eye and not enough fluid exiting the eye.
  • IOP intraocular pressure
  • Elevated IOP which can be measured by a "tonometry” test, results from fluid entering the eye and not enough fluid exiting the eye.
  • fluid enters the eye by seeping out of the blood vessels in the ciliary body. This fluid eventually makes its way past the crystalline lens, through the pupil (the central opening in the iris), and into the irido-corneal angle, the anatomical angle formed where the iris and the cornea come together. Then the fluid passes through the trabecular meshwork in the angle and leaves the eye via the canal of Schlemm.
  • Open angle glaucoma also can be caused when the posterior portion of the iris adheres to the anterior surface of the lens creating a "pupillary block", and preventing intraocular fluid from passing through the pupil into the anterior chamber.
  • Intraocular eye pressure is normal (between 12 - 22 mmHg) in about 25 - 30% glaucoma cases in the US, a condition known as normal-tension glaucoma. (In Japan, the rates may be as high as 70%.) Other factors are present that cause optic nerve damage but do not affect IOP.
  • Closed-angle glaucoma (also called angle-closure glaucoma) is responsible for 15% of all glaucoma cases. It is less common than POAG in the U.S., but it constitutes about half of the world's glaucoma cases because of its higher prevalence among Asians. The iris is pushed against the lens, sometimes sticking to it, closing off the drainage angle. This can occur very suddenly, resulting in an immediate rise in pressure. It often occurs in genetically susceptible people when the pupil shrinks suddenly. Closed-angle glaucoma can also be chronic and gradual, a less common condition.
  • Congenital glaucoma in which the eye's drainage canals fail to develop correctly, is present from birth. It is very rare, occurring in about 1 in 10,000 newborns. This may be an inherited condition and often can be corrected with microsurgery.
  • the present invention provides a method of attenuating expression of a target ocular mRNA in the eye of a subject suffering from glaucoma, comprising topically (non- invasively) administering to the surface of the eye of the subject an effective amount of at least one chemically modified double stranded RNA and a pharmaceutically acceptable carrier.
  • the at least one ocular target mRNA is a product of a gene selected from a list in Table A set forth in SEQ ID NOS: 1-293.
  • the target ocular mRNA is a product of a gene selected from CASP2, ASPP1, TP53BP2, BNIP3, RTP801L, ACHE, ADRB1 and CAPNS1.
  • the double stranded RNA is formulated for delivery as ear drops.
  • Neurodegenerative diseases are conditions in which cells of the CNS (the brain and / or the spinal cord and/or the eye) are lost.
  • the CNS cells are not readily regenerated en masse, so excessive damage can be devastating.
  • Neurodegenerative diseases result from deterioration of neurons or their myelin sheath, which over time leads to dysfunction and disabilities. They are crudely divided into two groups according to phenotypic effects, although these are not mutually exclusive: conditions affecting movement, such as ataxia; and conditions affecting memory and related to dementia. Dementia is marked by loss of intellectual functions such as memory, learning, reasoning, problem solving, and abstract thinking while vegetative functions remain intact.
  • Non- limiting examples of neurodegenerative disease are Alzheimer's disease, Amyotrophic lateral sclerosis (ALS, also known as Lou Gehrig's Disease), Huntington's disease, Lewy body dementia and Parkinson's disease.
  • CJD Creutzfeldt- Jakob disease
  • variant CJD Melt- Jakob disease
  • Non-limiting examples of ocular neurodegenerative disease include photoreceptor loss in the retina in subjects afflicted with macular degeneration, diabetic retinopathy, retinitis pigmentosa, glaucoma, and similar diseases.
  • the otic pharmaceutical compositions of the invention are useful for treating neurodegenerative diseases and conditions.
  • the otic pharmaceutical compositions of the present invention are particularly useful in treating a subject suffering from or affected by or susceptible to neurodegenerative disorders, including, without being limited to, Parkinson's disease, Amyotrophic Lateral Sclerosis (ALS), Prion disease dementia, Alzheimer's disease, Lewy body dementia, Pick's disease, Ataxia- telangiectasia (AT), Fro nto temporal dementia (FTD), Fro nto temporal lobar degeneration (FTLD), Huntington's disease, HIV-associated dementia, post-stroke dementia or any other disease- induced dementia; and ocular neurodegenerative diseases.
  • Parkinson's disease Amyotrophic Lateral Sclerosis
  • Prion disease dementia Alzheimer's disease
  • Lewy body dementia Lewy body dementia
  • Pick's disease Ataxia- telangiectasia
  • FDD Fro nto temporal dementia
  • FTLD Fro nto temporal lobar degeneration
  • Huntington's disease HIV-associated dementia
  • post-stroke dementia post-stroke dementia or any other
  • AD Alzheimer's Disease
  • the neurodegenerative disorder is Alzheimer's disease (AD).
  • AD is progressive, neurodegenerative disease characterized by loss of function and death of nerve cells in several areas of the brain leading to loss of cognitive function such as memory and language.
  • the present invention further provides a method of treating AD in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one gene expressed in the CNS of the subject in an amount effective to treat AD.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme, or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one target gene expressed in the CNS of the subject and associated with an AD. In certain embodiments inhibition of at least one target gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from an AD.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is selected from APP (SEQ ID NO: l- 5), BACE1 (SEQ ID NO: 13- 16), ADRB1 (SEQ ID NO:31), CDK5R1 (SEQ ID NO:35), MAPT (SEQ ID NO:6-l l), CASP3 (SEQ ID NO:24-25), TGM2 (SEQ ID NO:28-29), CAMK2A (SEQ ID NO:32-33), GABRA1 (SEQ ID NO:36-42), SYT1 (SEQ ID NO:63-65) and CASP2 (SEQ ID NO: 22-23).
  • APP SEQ ID NO: l- 5
  • BACE1 SEQ ID NO: 13- 16
  • ADRB1 SEQ ID NO:31
  • CDK5R1 SEQ ID NO:35
  • MAPT SEQ ID NO:6-l l
  • CASP3 SEQ ID NO:24-25
  • TGM2 SEQ ID NO:28-29
  • CAMK2A SEQ ID NO:32-33
  • the neurodegenerative disorder is Amyotrophic Lateral Sclerosis (ALS).
  • ALS a progressive, usually fatal, neurodegenerative disease caused by the degeneration of motor neurons, the nerve cells in the central nervous system that control voluntary muscle movement.
  • the disorder causes muscle weakness and atrophy throughout the body as both the upper and lower motor neurons degenerate, ceasing to send messages to muscles.
  • Unable to function the muscles gradually weaken, develop fasciculations (twitches) because of denervation, and eventually atrophy because of that denervation.
  • Subject suffering from ALS may ultimately lose the ability to initiate and control all voluntary movement; bladder and bowel sphincters and the muscles responsible for eye movement are usually (but not always) spared.
  • the present invention further provides a method of treating ALS in a subject in need of treatment that comprises administering to canal of the subject's ear an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat ALS.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified siRNA, a chemically modified siRNA, a shRNA, an aptamer, a ribozyme, a dsRNA or DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified siRNA inhibits expression of at least one target gene expressed in the CNS of the subject and associated with ALS. In certain embodiments inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from an ALS.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is selected from SOD1 (SEQ ID NO: 12), TARDBP (SEQ ID NO:30), CBLN1 (SEQ ID NO:34), CDK5R1 (SEQ ID NO:35), FUS (SEQ ID NO:67-70) and CASP2 (SEQ ID NO:22-23).
  • the neurodegenerative disorder is Parkinson's Disease (PD).
  • Parkinson's disease is a progressive disorder of the nervous system marked by muscle tremors, muscle rigidity, decreased mobility, stooped posture, slow voluntary movements, and a mask-like facial expression.
  • the present invention further provides a method of treating PD in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat PD.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with PD.
  • inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from PD.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is selected from MAPT (SEQ ID NO:4-7), NPTX2 (SEQ ID NO:26), SYT1 (SEQ ID NO:32), MAPK10 (SEQ ID NO:24), NTS (SEQ ID NO:28), PDCD2 (SEQ ID NO:29), PENK (SEQ ID NO:31), CBLNl (SEQ ID NO:21) and CASP2 (SEQ ID NO: 22-23) .
  • Ataxia-telangiectasia (AT)
  • the neurodegenerative disorder is Ataxia- telangiectasia (AT).
  • AT is a rare, neurodegenerative, inherited disease which affects many parts of the body and causes severe disability. Ataxia refers to poor coordination and telangiectasia to small, dilated blood vessels, both of which are hallmarks of the disease. AT affects the cerebellum (the body's motor coordination control center) and also weakens the immune system in about 70% of the cases, leading to respiratory disorders and increased risk of cancer.
  • the present invention further provides a method of treating AT in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one gene expressed in the CNS of the subject in an amount effective to treat AT.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with AT. In certain embodiments inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from AT.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is CBLN1 (SEQ ID NO:21).
  • FTLD Frontotemporal Lobar Degeneration
  • the neurodegenerative disorder is Frontotemporal lobar degeneration (FTLD).
  • FTLD Frontotemporal lobar degeneration
  • FTLD is the name for a group of clinically, pathologically and genetically heterogeneous disorders associated with atrophy in the frontal lobe and temporal lobe of the brain, with sparing of the parietal and occipital lobes.
  • FTLD is probably the fourth most common cause of dementia after Alzheimer's disease, Dementia with Lewy bodies and vascular dementia.
  • it is the second most common cause after Alzheimer's disease.
  • FTLD frontotemporal dementia
  • semantic dementia progressive nonfluent aphasia
  • FTLD progressive nonfluent aphasia
  • AD Alzheimer's disease
  • MND motor neuron disease
  • the present invention further provides a method of treating FTLD in a subject in need of treatment that comprises administering to the canal of the subject's ear an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat FTLD.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with FTLD. In certain embodiments inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from FTLD.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is selected from MAPT (SEQ ID NO:4-7) and TARDBP (SEQ ID NO: 18).
  • the neurodegenerative disorder is Frontotemporal degeneration (FTD).
  • FTD is a non- Alzheimer dementia that may rank as the second most common cause of early onset dementia. Both behavioral and language presentations of FTD usually have earlier onset (mean onset in sixth decade of life) than Alzheimer disease (AD) (mean onset in eighth decade of life).
  • the present invention further provides a method of treating FTD in a subject in need of treatment that comprises administering to the canal of the subject's ear an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat FTD.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with FTD. In certain embodiments inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from FTD.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is NPTX2 (SEQ ID NO:26).
  • the neurodegenerative disorder is Pick's disease (PiD).
  • PiD is a rare neurodegenerative disease that is sometimes familial. Numerous different areas of the brain are affected by PiD, but the specific areas that are affected allow for differentiation between PiD and Alzheimer's disease.
  • Pick's disease (the pathology) causes progressive destruction of nerve cells in the brain and causes tau proteins to accumulate into "Pick bodies" that are a defining characteristic of the disease.
  • Pick's disease is one of the causes of the clinical syndrome of frontotemporal lobar degeneration which has three subtypes.
  • Pick's disease pathology is associated more with the frontotemporal dementia and progressive nonfluent aphasia subtypes than the semantic dementia subtype.
  • PiD has several unique biochemical characteristics that allow for unique identification of Pick's disease as opposed to other pathological subtypes of frontotemporal lobar degeneration.
  • Clinical features include aphasia; apraxia; confusion; anomia; memory loss; and personality deterioration. This pattern is consistent with the pathologic findings of circumscribed atrophy of the poles of the frontal lobe and temporal lobe.
  • Neuronal loss is maximal in the hippocampus, entorhinal cortex, and amygdala.
  • Some ballooned cortical neurons contain argentophylic (Pick) bodies (for further details see Brain Pathol 1998 Apr;8(2):339-54 and Adams et al., Principles of Neurology, 6th ed, ppl057-9).
  • the present invention further provides a method of treating PiD in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat PiD.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with PiD. In certain embodiments inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from PiD.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is MAPT (SEQ ID NO:6-l 1).
  • the neurodegenerative disorder is Huntington's disease (HD).
  • HD incurable, genetic neurodegenerative disorder.
  • Characteristic symptoms which are progressive and begin subtly, include uncoordinated, jerky body movements, decline in mental abilities and behavioral and psychiatric problems. Dementia is the norm as the disease advances.
  • the present invention further provides a method of treating HD in a subject in need of treatment that comprises administering to the canal of the subject's ear an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat HD.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with HD. In certain embodiments inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from HD.
  • the at least one target mRNA is selected from an mRNA polynucleotide HTT (SEQ ID NO: 168), TGM2 (SEQ ID NO:28-29) and ADRB1 (SEQ ID NO:31) .
  • the neurodegenerative disorder is Post Stroke Dementia (PSD). About 25% of people have dementia after a stroke with many others developing dementia over the following 5 to 10 years. In addition, many individuals experience more subtle impairments of their higher brain functions (such as planning skills and speed of processing information) and are at very high risk of subsequently developing dementia. Very small strokes in the deep parts of the brain in this process (called microvascular disease) seem to be essential in the process leading to an identified pattern of brain atrophy specific to post- stroke dementia.
  • PSD Post Stroke Dementia
  • the present invention further provides a method of treating PSD in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat PSD.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with PSD. In certain embodiments inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from PSD.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the neurodegenerative disease is a neurodegenerative disease of the eye, including photoreceptor loss in the retina in subjects afflicted with macular degeneration, diabetic retinopathy, retinitis pigmentosa, glaucoma, and similar diseases.
  • the present invention further provides a method of treating an ocular neurodegenerative disease in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat an ocular neurodegenerative disease.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with the ocular neurodegenerative disease. In certain embodiments inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from ocular neurodegenerative disease.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is CASP2 (SEQ ID NO:22-23).
  • the otic pharmaceutical compositions of the invention are useful for treating injury of the central nervous system (CNS).
  • the otic pharmaceutical compositions of the present invention are particularly useful in treating a subject suffering from or affected by or susceptible to injury of the CNS, including, without being limited to, traumatic and non-traumatic spinal cord injury, and brain injury (e.g. Traumatic Brain Injury (TBI)), that is caused by fracture or penetration of the skull (i.e. a vehicle accident, fall, gunshot wound), a disease process (i.e. neurotoxins, infections, tumors, metabolic abnormalities, etc.) or a closed head injury such as in the case of rapid acceleration or deceleration of the head (i.e. Shaken Baby Syndrome, blast), blunt trauma, concussions, and concussion syndrome.
  • TBI Traumatic Brain Injury
  • an ischemic episode may be caused by a mechanical injury to the Central Nervous System, such as results from a blow to the head or spine.
  • Trauma can involve a tissue insult such as an abrasion, incision, contusion, puncture, compression, etc., such as can arise from traumatic contact of a foreign object with any locus of or appurtenant to the head, neck, or vertebral column.
  • Other forms of traumatic injury can arise from constriction or compression of CNS tissue by an inappropriate accumulation of fluid (for example, a blockade or dysfunction of normal cerebrospinal fluid or vitreous humor fluid production, turnover, or volume regulation, or a subdural or intracranial hematoma or edema).
  • traumatic constriction or compression can arise from the presence of a mass of abnormal tissue, such as a metastatic or primary tumor.
  • the injury to the CNS is Spinal Cord Injury (SCI) or myelopathy.
  • SCI or myelopathy is a disturbance of the spinal cord that results in loss of sensation and/or mobility.
  • the two common types of spinal cord injury are due to trauma and disease. Traumatic injury can be due to automobile accidents, falls, gunshot, diving accidents inter alia, and diseases that can affect the spinal cord include polio, spina bifida, tumors, Amyotrophic Lateral Sclerosis (ALS), Multiple Sclerosis (MS) syringomyelia, transverse myelitis and Friedreich' s ataxia.
  • ALS Amyotrophic Lateral Sclerosis
  • MS Multiple Sclerosis
  • the otic pharmaceutical compositions of the invention are used for treating or preventing the damage caused by spinal-cord injury especially spinal cord trauma caused by motor vehicle accidents, falls, sports injuries, industrial accidents, gunshot wounds, spinal cord trauma caused by spine weakening (such as from rheumatoid arthritis or osteoporosis) or if the spinal canal protecting the spinal cord has become too narrow (spinal stenosis) due to the normal aging process, direct damage that occur when the spinal cord is pulled, pressed sideways, or compressed, damage to the spinal-cord following bleeding, fluid accumulation, and swelling inside the spinal cord or outside the spinal cord (but within the spinal canal).
  • the otic pharmaceutical compositions of the invention are also used for treating or preventing the damage caused by spinal-cord injury due to disease such as polio or spina bifida.
  • the present invention further provides a method of treating SCI in a subject in need of treatment that comprises administering to the canal of the subject's ear an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat SCI.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with SCI. In certain embodiments inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from SCI.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is selected from CASP3 (SEQ ID NO:24-25), TGM2 (SEQ ID NO:28-29), CAMK2A (SEQ ID NO:32-33), CBLN1 (SEQ ID NO:34), CDK5R1 (SEQ ID NO:35), MAPK10 (SEQ ID NO:43-46), NOS1 (SEQ ID NO:47), NTS (SEQ ID NO:51) and CASP2 (SEQ ID NO: 22-23).
  • CASP3 SEQ ID NO:24-25
  • TGM2 SEQ ID NO:28-29
  • CAMK2A SEQ ID NO:32-33
  • CBLN1 SEQ ID NO:34
  • CDK5R1 SEQ ID NO:35
  • MAPK10 SEQ ID NO:43-46
  • NOS1 SEQ ID NO:47
  • NTS SEQ ID NO:51
  • CASP2 SEQ ID NO: 22-23
  • the injury to the CNS is brain injury.
  • Brain injury such as trauma and stroke are among the leading causes of mortality and disability in the western world.
  • Traumatic brain injury is one of the most serious reasons for hospital admission and disability in modern society. Clinical experience suggests that TBI may be classified into primary damage occurring immediately after injury, and secondary damage, which occurs during several days post injury. Current therapy of TBI is either surgical or else mainly symptomatic.
  • the present invention further provides a method of treating brain injury in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat brain injury.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with brain injury. In certain embodiments inhibition of at least one gene confers upon the CNS neuroprotective properties.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from brain injury.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the otic pharmaceutical compositions of the invention are useful for treating neoplasms in the central nervous system (CNS).
  • the neoplasm in the CNS is selected from any intracranial tumor created by abnormal and uncontrolled cell division either in the brain itself or spread from cancers primarily located in other organs (i.e. metastatic tumors).
  • the neoplasm in the CNS is created by abnormal proliferation of or in the, inter alia, neurons (e.g. Motor neuron, Purkinje neuron, GABAergic neuron, Multipolar neuron, Cerebellar neuron, Afferent neuron, Sensory neuron), glial cells (e.g.
  • neurons e.g. Motor neuron, Purkinje neuron, GABAergic neuron, Multipolar neuron, Cerebellar neuron, Afferent neuron, Sensory neuron
  • glial cells e.g.
  • astrocytes astrocytes, microglia, oligodendrocytes), ependymal cells, lymphatic tissue, blood vessels, cranial nerves, myelin-producing Schwann cells, meninges, skull, Striatum, Nucleus of stria terminalis, hypothalamus, pituitary gland and pineal gland.
  • otic pharmaceutical compositions of the present invention are particularly useful in treating a subject suffering from or affected by or susceptible to intracranial glioma selected from, without being limited to, ependymoma, glioma, astrocytoma, oligodendroglioma and oligoastrocytoma.
  • otic pharmaceutical compositions of the present invention are useful in treating a subject suffering Pilocytic astrocytoma of cerebellum or Oligodendroglioma of brain.
  • the neoplasm is a neural crest tumor such as e.g. cranial primitive neuroectodermal tumors (PNET).
  • PNET cranial primitive neuroectodermal tumors
  • the otic pharmaceutical compositions of the present invention are useful in treating a subject suffering from a neoplasm selected from, without being limited to, Medulloblastoma of cerebellum, Neuroblastoma of brain, Glioblastoma multiforme of brain and Neurofibromatosis.
  • the neoplasm in the CNS is intracranial glioma (e.g. Pilocytic astrocytoma of cerebellum; Oligodendroglioma of brain; Glioblastoma multiforme of brain; Astrocytoma).
  • intracranial glioma e.g. Pilocytic astrocytoma of cerebellum; Oligodendroglioma of brain; Glioblastoma multiforme of brain; Astrocytoma.
  • Astrocytoma is a glial tumor of the brain or spinal cord showing astrocytic differentiation. It includes the following clinicopathological entities: pilocytic astrocytoma, diffuse astrocytoma, anaplastic astrocytoma, pleomorphic xanthoastrocytoma, subependymal giant cell astrocytoma, and glioblastoma.
  • the present invention further provides a method of treating astrocytoma in a subject in need of treatment that comprises administering to the canal of the subject's ear an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat astrocytoma.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with astrocytoma.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is selected from CAMK2A (SEQ ID NO:32-33), CBLN1 (SEQ ID NO:34), SYT1 (SEQ ID NO:63-65) and GABRA1 (SEQ ID NO:36-42).
  • the neoplasm is pilocytic astrocytoma of cerebellum - a WHO Grade 1 astrocytoma which arises in the cerebellum.
  • the tumor is composed of spindle shaped cells with numerous collections of reddish astrocytic fibers called Rosenthal fibers.
  • Rosenthal fibers Over 80% or the cerebellar astrocytomas of childhood are pilocytic.
  • Pilocytic astrocytomas may rarely occur in adults. They are usually treated by surgical resection and in most cases have a favorable prognosis.
  • the present invention further provides a method of treating pilocytic astrocytoma of cerebellum in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat pilocytic astrocytoma of cerebellum.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with pilocytic astrocytoma of cerebellum.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from pilocytic astrocytoma of cerebellum.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is selected from GABRA1 (SEQ ID NO:36-42), NOS1 (SEQ ID NO:47) and NRGN (SEQ ID NO:49- 50).
  • the neoplasm is Neurofibromatosis.
  • Neurofibromatosis is group of disorders characterized by an autosomal dominant pattern of inheritance with high rates of spontaneous mutation and multiple neurofibromas or neurilemmomas; neurofibromatosis 1 (generalized neurofibromatosis) accounts for approximately 95% of cases, although multiple additional subtypes (e.g., neurofibromatosis 2, neurofibromatosis 3, etc.) have been described.
  • the present invention further provides a method of treating Neurofibromatosis in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat Neurofibromatosis.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the brain of the subject and associated with Neurofibromatosis.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from Neurofibromatosis.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the spinal cord of the subject.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is NOS1 (SEQ ID NO:47).
  • the otic pharmaceutical compositions of the invention are useful for treating neurological disorders.
  • the neurological disorder is selected from, without being limited to, stroke, stroke-like situations (e.g. cerebral, renal, cardiac failure), neuronal cell death, epilepsy, Parkinsonism, Gluten Ataxia, cerebral ischemia and cerebrovascular accident.
  • the neurological disorder is epilepsy.
  • Epilepsy is a group of disorders marked by problems in the normal functioning of the brain. These problems can produce seizures, unusual body movements, loss of consciousness or changes in consciousness, as well as mental problems or problems with the senses.
  • the present invention further provides a method of treating epilepsy in a subject in need of treatment that comprises administering to the canal of the subject's ear an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat epilepsy.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with epilepsy.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from epilepsy.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the spinal cord of the subject.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is selected from GABRA1 (SEQ ID NO:36-42) and MAPK10 (SEQ ID NO:43-46).
  • the neurological disease is cerebrovascular disorder
  • Cerebrovascular accident is a sudden, nonconvulsive loss of neurological function due to an ischemic or hemorrhagic intracranial vascular event.
  • cerebrovascular accidents are classified by anatomic location in the brain, vascular distribution, etiology, age of the affected individual, and hemorrhagic vs. nonhemorrhagic nature (for additional information see Adams et al., Principles of Neurology, 6th ed, pp777-810).
  • Cerebrovascular diseases occur predominately in the middle and late years of life. They cause approximately 200,000 deaths in the United States each year as well as considerable neurological disability. The incidence of stroke increases with age and affects many elderly people, a rapidly growing segment of the population. These diseases cause either ischemia- infarction or intracranial hemorrhage. Stroke
  • the neurological disorder is stroke.
  • Stroke is an acute neurological injury occurring as a result of interrupted blood supply, resulting in an insult to the brain.
  • Most cerebrovascular diseases present as the abrupt onset of focal neurological deficit. The deficit may remain fixed, or it may improve or progressively worsen, leading usually to irreversible neuronal damage at the core of the ischemic focus, whereas neuronal dysfunction in the penumbra may be treatable and/or reversible.
  • Prolonged periods of ischemia result in frank tissue necrosis. Cerebral edema follows and progresses over the subsequent 2 to 4 days. If the region of the infarction is large, the edema may produce considerable mass effect with all of its attendant consequences. Damage to neuronal tissue can lead to severe disability and death.
  • the extent of the damage is primarily affected by the location and extent of the injured tissue. Endogenous cascades activated in response to the acute insult play a role in the functional outcome. Efforts to minimize, limit and/or reverse the damage have the great potential of alleviating the clinical consequences.
  • the present invention further provides a method of treating cerebrovascular condition in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one gene expressed in the CNS of the subject in an amount effective to treat cerebrovascular condition.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with cerebrovascular condition.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from cerebrovascular condition.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is selected from MAPK10 (SEQ ID NO:43-46) and PDE4D (SEQ ID NO:58-60).
  • the neurological disorder is Parkinsonism - a group of disorders which feature impaired motor control characterized by bradykinesia, muscle rigidity; tremor; and postural instability.
  • Parkinsonian diseases are generally divided into primary parkinsonism, secondary parkinsonism and inherited forms. These conditions are associated with dysfunction of dopaminergic or closely related motor integration neuronal pathways in the basal ganglia.
  • the present invention further provides a method of treating parkinsonism in a subject in need of treatment that comprises administering to the canal of the subject's ear an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat parkinsonism.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with parkinsonism.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from parkinsonism.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is MAPT (SEQ ID NO:4-7).
  • the neurological disorder is Gluten Ataxia (GA) - cerebellar dysfunction caused by sensitivity to gluten.
  • GA Gluten Ataxia
  • the present invention further provides a method of treating GA in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat GA.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with GA.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from GA.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is TGM2 (SEQ ID NO: 15- 16).
  • the otic pharmaceutical compositions of the invention are useful for treating mood disorders (e.g. major depressive disorder and bipolar disorder) and Posttraumatic stress disorder.
  • diagnosticians recognize several subtypes of major depressive disorders, such as for example, melancholic depression, psychotic depression, catatonic depression, postpartum depression, seasonal affective disorder.
  • Dysthymia is a chronic, milder mood disturbance where a person reports a depressed mood almost daily over a span of at least two years. Dysthymic patients are vulnerable to secondary episodes of major depression (sometimes referred to as double depression).
  • Depressive Disorder Not Otherwise Specified is a classification used for depressive disorders that are impairing but do not fit any of the officially specified diagnoses. It includes the research diagnoses of Recurrent brief depression, and Minor Depressive Disorder.
  • Recurrent brief depression (RBD) is distinguished from Major Depressive Disorder primarily by differences in duration. People with RBD have depressive episodes about once per month, with individual episodes lasting less than two weeks and typically less than 2-3 days.
  • Minor Depression refers to a depression that does not meet full criteria for major depression but in which at least two symptoms are present for two weeks. In one embodiment the mood disorder is major depressive disorder.
  • the present invention further provides a method of treating major depressive disorder in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat major depressive disorder.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated major depressive disorder.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from major depressive disorder.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is NRGN (SEQ ID NO:27).
  • the mood disorder is bipolar disorder.
  • the present invention further provides a method of treating bipolar disorder in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat bipolar disorder.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with bipolar disorder.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from bipolar disorder.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is SYT1 (SEQ ID NO:32).
  • the CNS disorder is post-traumatic stress disorder - acute, chronic, or delayed reactions to traumatic events such as military combat, assault, or natural disaster.
  • the present invention further provides a method of post-traumatic stress disorder in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat post- traumatic stress disorder.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with post-traumatic stress disorder.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from post-traumatic stress disorder.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is NRGN (SEQ ID NO:27).
  • the otic pharmaceutical compositions of the invention are useful in treating disease or disorder of the CNS, selected from, without being limited to, Supranuclear paralysis, Lymphocytic choriomeningitis, Niemann Pick disease (e.g. Niemann Pick disease Type C) and AF type amyloidosis (Familial neuropathic amyloidosis). Lymphocytic Choriomeningitis
  • the CNS disease is Lymphocytic choriomeningitis - benign viral infection of meninges and central nervous system producing an infiltration of lymphocytes in the choroid plexus.
  • the present invention further provides a method of treating Lymphocytic choriomeningitis in a subject in need of treatment that comprises administering to the canal of the subject's ear an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat Lymphocytic choriomeningitis.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, an shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated Lymphocytic chorio meningitis .
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from Lymphocytic choriomeningitis.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is CASP3 (SEQ ID NO: 13- 14).
  • the CNS disorder is AF type amyloidosis - a group of inherited disorders of the peripheral nervous system associated with the deposition of amyloid in nerve tissue.
  • the different clinical types based on symptoms correspond to the presence of a variety of mutations in several different proteins including Transthyretin (Prealbumin); Apolipoprotein A-I; and Gelsolin.
  • the present invention further provides a method of treating AF type amyloidosis in a subject in need of treatment that comprises administering to the ear of the subject an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to treat AF type amyloidosis.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with AF type amyloidosis.
  • the present invention provides a method of attenuating expression of a target mRNA in a subject suffering from AF type amyloidosis.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the gene is TTR (SEQ ID NO:33).
  • the otic pharmaceutical compositions of the invention are directed to providing neuroprotection, or to provide cerebroprotection, and to attenuating acute or chronic neuronal damage in diseases, disorders or injury of the CNS.
  • the present invention further provides a method of conferring neuroprotection to cells and/or tissues in a subject in need of treatment that comprises administering to the canal of the subject's ear an otic pharmaceutical composition which comprises a therapeutically effective amount of at least one oligonucleotide compound, which inhibits expression of at least one target gene expressed in the CNS of the subject in an amount effective to confer upon the cells of the CNS neuroprotective properties.
  • the oligonucleotide compound is selected from the group consisting of an antisense, an unmodified double stranded RNA, a chemically modified double stranded RNA, a shRNA, an aptamer, a ribozyme or a DNA compound.
  • the oligonucleotide compound is chemically modified siRNA.
  • the chemically modified double stranded RNA inhibits expression of at least one gene expressed in the CNS of the subject and associated with an acute or a chronic neuronal damage in diseases, disorders or injury of the CNS.
  • the present invention provides a method of a method of conferring neuroprotection to cells and/or tissues in a subject in need of treatment.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the siRNA targets RhoA (SEQ ID NO: 110), APP (SEQ ID NOS: 1-5), CASP2 (SEQ ID NOS:22-23), FAS (SEQ ID NOS: 1524-160), FASLG (SEQ ID NO: 161), NOS1 (SEQ ID NO:47).
  • the present invention provides a method of a method of reducing neurotoxicity in cells and/or tissues in a subject in need of treatment.
  • the at least one target mRNA is selected from an mRNA polynucleotide set forth in any one of SEQ ID NOS: 1-293.
  • the siRNA targets RhoA (SEQ ID NO: 110), APP (SEQ ID NOS: 1-5), CASP2 (SEQ ID NOS:22-23), FAS (SEQ ID NOS: 1524-160), FASLG (SEQ ID NO: 161), NOS1 (SEQ ID NO:47).
  • PCR Polymerase chain reaction
  • FACS Flow Cytometry
  • ELISA is a preferred immunoassay.
  • ELISA assays are well known to those skilled in the art. Both polyclonal and monoclonal antibodies can be used in the assays. Where appropriate other immunoassays, such as radioimmunoassays (RIA) can be used as are known to those in the art. Available immunoassays are extensively described in the patent and scientific literature. See, for example, United States Patent Nos.
  • siRNA compounds are transfected with siRNA compounds using the LipofectamineTM 2000 reagent (Invitrogen) at final concentrations of 5nM or 20nM. The cells are incubated at 37°C in a C0 2 incubator for 72h. As positive control for transfection PTEN-Cy3 labeled siRNA compounds are used. Various chemically modified siRNA compounds are tested for activity. GFP siRNA compounds are used as negative control for siRNA activity.
  • the percent of inhibition of gene expression using specific preferred siRNA structures is determined using qPCR analysis of a target gene in cells expressing the endogenous gene.
  • siRNAs having specific sequences that are selected for in vitro testing are specific for human and a second species such as non-human primate, rat or rabbit genes.
  • Chemically modified siRNA compounds according to the present invention are tested for duplex stability in human serum, as follows:
  • siRNA molecules at final concentration of 7uM are incubated at 37°C in 100% human serum (Sigma Cat# H4522). (siRNA stock lOOuM diluted in human serum 1: 14.29).
  • Example 1 Non- Invasive (Ear Drops) Delivery of siRNA Comprising Otic Pharmaceutical Compositions to the CNS in Mice Assessed by Stem - Loop qPCR
  • Study design The study included 8 experimental groups with 6 mice each as described in Table 1. Animals from experimental groups I-IV: were treated with a single dose of otic pharmaceutical composition comprising siRNA compound that targets the mRNA of the CASP2 gene. Table 1: Study Design

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Ophthalmology & Optometry (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne des méthodes non invasives de traitement de maladies, d'affections et de lésions du système nerveux central (SNC) et, en particulier, des compositions otiques et leurs procédés d'utilisation.
PCT/US2010/059597 2009-12-09 2010-12-09 Méthodes et compositions utilisées pour le traitement de maladies, d'affections ou de lésions du snc WO2011072091A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US13/514,880 US8778904B2 (en) 2009-12-09 2010-12-09 Methods and compositions for treating diseases, disorders or injury of the CNS
EP10809113.3A EP2510098B1 (fr) 2009-12-09 2010-12-09 Méthodes et compositions utilisées pour le traitement de maladies, d'affections ou de lésions du snc

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US26783509P 2009-12-09 2009-12-09
US61/267,835 2009-12-09
US31989410P 2010-04-01 2010-04-01
US61/319,894 2010-04-01

Publications (1)

Publication Number Publication Date
WO2011072091A1 true WO2011072091A1 (fr) 2011-06-16

Family

ID=44145908

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/059597 WO2011072091A1 (fr) 2009-12-09 2010-12-09 Méthodes et compositions utilisées pour le traitement de maladies, d'affections ou de lésions du snc

Country Status (3)

Country Link
US (1) US8778904B2 (fr)
EP (2) EP2510098B1 (fr)
WO (1) WO2011072091A1 (fr)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013037499A1 (fr) * 2011-09-15 2013-03-21 Johann Wolfgang Goethe-Universität Inhibiteurs de l'expression de nox4 et/ou de la fonction de nox4 et leur utilisation pour la prévention et le traitement des lésions nerveuses et/ou de la douleur neuropathique
WO2013067076A2 (fr) 2011-11-03 2013-05-10 Quark Pharmaceuticals, Inc. Procédés et compositions pour la neuroprotection
WO2013070821A1 (fr) 2011-11-08 2013-05-16 Quark Pharmaceuticals, Inc. Méthodes et compositions destinées à traiter des maladies, des troubles ou une lésion du système nerveux
WO2013103632A1 (fr) * 2012-01-04 2013-07-11 Quark Pharmaceuticals, Inc. Composés arn double brin dirigés contre casp2 et leurs utilisations
WO2015006475A1 (fr) * 2013-07-12 2015-01-15 Biogen Idec International Neuroscience Gmbh Biomarqueurs génétiques et d'imagerie associés au déclin des mesures cognitives et du métabolisme cérébral du glucose chez des sujets souffrant de la maladie d'alzheimer ou risquant de la développer
CN104357470A (zh) * 2014-11-14 2015-02-18 四川大学华西第二医院 一种HtrA2融合基因及其应用
WO2016029191A3 (fr) * 2014-08-22 2016-08-18 Auckland Uniservices Limited Modulateurs de canaux
CN106967789A (zh) * 2017-01-26 2017-07-21 上海长海医院 一种前列腺癌标志物plxna1及其应用
US9828420B2 (en) 2007-01-05 2017-11-28 University Of Zürich Method of providing disease-specific binding molecules and targets
US10842871B2 (en) 2014-12-02 2020-11-24 Biogen International Neuroscience Gmbh Methods for treating Alzheimer's disease
CN112716971A (zh) * 2021-01-19 2021-04-30 苏州大学 lncRNA XR_595534.2在制备治疗或预防慢性疼痛的药物中的应用
EP4035659A1 (fr) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes destinés à l'administration d'agents thérapeutiques
US11655289B2 (en) 2017-08-22 2023-05-23 Biogen Ma Inc. Pharmaceutical compositions containing anti-beta amyloid antibodies

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8614311B2 (en) * 2007-12-12 2013-12-24 Quark Pharmaceuticals, Inc. RTP801L siRNA compounds and methods of use thereof
FR2930447B1 (fr) * 2008-04-25 2010-07-30 Sod Conseils Rech Applic Utilisation therapeutique d'au moins une neurotoxine botulique dans le traitement de la douleur dans le cas de la neuropathie diabetique
EP3027223A1 (fr) 2013-07-31 2016-06-08 QBI Enterprises Ltd. Procédés d'utilisation de composés sphingolipide-polyalkylamine-oligonucléotide
US20180104309A1 (en) * 2015-05-11 2018-04-19 Duke University Compositions and methods for spinal cord regeneration
EP3400003B1 (fr) * 2016-01-05 2023-05-03 The Trustees of Columbia University in the City of New York Compositions pour la régulation de l'activité de l'inhibiteur de la protéine de liaison à l'adn de type 2 (id2)
CA3124090A1 (fr) * 2018-12-19 2020-06-25 Alnylam Pharmaceuticals, Inc. Compositions d'agent d'arni de precurseur de la proteine amyloide (app) et leurs methodes d'utilisation
WO2023023189A2 (fr) * 2021-08-17 2023-02-23 University Of Virginia Patent Foundation Surexpression de la clustérine dans la maladie d'alzheimer
WO2023129947A2 (fr) * 2021-12-29 2023-07-06 Salk Institute For Biological Studies Méthodes de réduction de la glycémie et de traitement du diabète de type 2 par activation de pde4d3

Citations (76)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3791932A (en) 1971-02-10 1974-02-12 Akzona Inc Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3839153A (en) 1970-12-28 1974-10-01 Akzona Inc Process for the detection and determination of specific binding proteins and their corresponding bindable substances
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3850578A (en) 1973-03-12 1974-11-26 H Mcconnell Process for assaying for biologically active molecules
US3853987A (en) 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
US3867517A (en) 1971-12-21 1975-02-18 Abbott Lab Direct radioimmunoassay for antigens and their antibodies
US3879262A (en) 1972-05-11 1975-04-22 Akzona Inc Detection and determination of haptens
US3901654A (en) 1971-06-21 1975-08-26 Biological Developments Receptor assays of biologically active compounds employing biologically specific receptors
US3935074A (en) 1973-12-17 1976-01-27 Syva Company Antibody steric hindrance immunoassay with two antibodies
US3984533A (en) 1975-11-13 1976-10-05 General Electric Company Electrophoretic method of detecting antigen-antibody reaction
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4034074A (en) 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US4098876A (en) 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
US4439196A (en) 1982-03-18 1984-03-27 Merck & Co., Inc. Osmotic drug delivery system
US4447224A (en) 1982-09-20 1984-05-08 Infusaid Corporation Variable flow implantable infusion apparatus
US4447233A (en) 1981-04-10 1984-05-08 Parker-Hannifin Corporation Medication infusion pump
US4475196A (en) 1981-03-06 1984-10-02 Zor Clair G Instrument for locating faults in aircraft passenger reading light and attendant call control system
US4486194A (en) 1983-06-08 1984-12-04 James Ferrara Therapeutic device for administering medicaments through the skin
US4487603A (en) 1982-11-26 1984-12-11 Cordis Corporation Implantable microinfusion pump system
US4666828A (en) 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4801531A (en) 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
US4879219A (en) 1980-09-19 1989-11-07 General Hospital Corporation Immunoassay utilizing monoclonal high affinity IgM antibodies
US4925678A (en) 1987-04-01 1990-05-15 Ranney David F Endothelial envelopment drug carriers
US4959217A (en) 1986-05-22 1990-09-25 Syntex (U.S.A.) Inc. Delayed/sustained release of macromolecules
US5011771A (en) 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US5167616A (en) 1989-12-14 1992-12-01 Alza Corporation Iontophoretic delivery method
US5169383A (en) 1988-10-03 1992-12-08 Alza Corporation Control membrane for electrotransport drug delivery
US5192659A (en) 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5225182A (en) 1991-10-31 1993-07-06 Sharma Yash P Vectored drug delivery system using a cephaloplastin linking agent and a methed of using the system
WO1993023569A1 (fr) 1992-05-11 1993-11-25 Ribozyme Pharmaceuticals, Inc. Procede et reactif d'inhibition de la replication virale
US5272057A (en) 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5281521A (en) 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
EP0586520A1 (fr) 1991-05-21 1994-03-16 Isis Pharmaceuticals, Inc. Analogues d'oligonucleotides a squelette modifie
US5580859A (en) 1989-03-21 1996-12-03 Vical Incorporated Delivery of exogenous DNA sequences in a mammal
US5593972A (en) 1993-01-26 1997-01-14 The Wistar Institute Genetic immunization
WO1998039352A1 (fr) 1997-03-07 1998-09-11 Takeshi Imanishi Nouveaux analogues de bicyclonucleoside et d'oligonucleotide
WO1999014226A2 (fr) 1997-09-12 1999-03-25 Exiqon A/S Analogues d'oligonucleotides
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
WO2000044895A1 (fr) 1999-01-30 2000-08-03 Roland Kreutzer Methode et medicament destines a inhiber l'expression d'un gene donne
WO2000044914A1 (fr) 1999-01-28 2000-08-03 Medical College Of Georgia Research Institute, Inc. Composition et methode destinees a l'attenuation in vivo et in vitro de l'expression genique utilisant de l'arn double brin
WO2000047599A1 (fr) 1999-02-12 2000-08-17 Sankyo Company, Limited Nouveaux analogues de nucleosides et d'oligonucleotides
WO2000049035A1 (fr) 1999-02-19 2000-08-24 The General Hospital Corporation Inhibition d'un gene
US6121426A (en) 1988-12-29 2000-09-19 Bio-Technology General Corp. Fibrin binding domain polypeptides and uses and methods of producing same
WO2000063364A2 (fr) 1999-04-21 2000-10-26 American Home Products Corporation Procedes et compositions pour l'inhibition de la fonction de sequences polynucleotidiques
WO2001029058A1 (fr) 1999-10-15 2001-04-26 University Of Massachusetts Genes de voies d'interference d'arn en tant qu'outils d'interference genetique ciblee
WO2001036646A1 (fr) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibition d"expression genique a l"aide d"arn bicatenaire
WO2001036641A2 (fr) 1999-11-02 2001-05-25 Chiron Corporation DETECTEURS DES RECEPTEURS D'ARN DOUBLE BRIN (dsRNA-R) ET PROCEDES CORRESPONDANTS
EP0618925B1 (fr) 1991-12-24 2001-08-29 Isis Pharmaceuticals, Inc. Oligonucleotides antisense
WO2001075164A2 (fr) 2000-03-30 2001-10-11 Whitehead Institute For Biomedical Research Mediateurs d'interference arn specifiques de sequences arn
WO2002044321A2 (fr) 2000-12-01 2002-06-06 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Petites molecules d'arn mediant l'interference arn
WO2002086105A1 (fr) 2001-04-20 2002-10-31 Chiron Corporation Apport d'agents polynucleotides au systeme nerveux central
US6586238B1 (en) 1996-04-16 2003-07-01 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids containing 5′-and or 3′-cap structures
US6602858B2 (en) 1997-03-31 2003-08-05 Ribozyme Pharmaceuticals, Inc. Nucleic acid catalysts comprising L-nucleotide analogs
WO2003087368A2 (fr) * 2002-04-18 2003-10-23 Lynkeus Bio Tech Gmbh Moyens et procedes pour la modulation specifique de genes cibles dans le snc et l'oeil, et procedes pour leur identification
US20040019001A1 (en) 2002-02-20 2004-01-29 Mcswiggen James A. RNA interference mediated inhibition of protein typrosine phosphatase-1B (PTP-1B) gene expression using short interfering RNA
WO2004015107A2 (fr) 2002-08-05 2004-02-19 Atugen Ag Nouvelles formes de molecules d'arn interferant
WO2004041924A2 (fr) 2002-11-05 2004-05-21 Isis Pharmaceuticals, Inc. Composes oligomeres a liaison non phosphore et leur utilisation pour la modulation genique
US20050004064A1 (en) 2001-11-21 2005-01-06 Mitsubishi Chemical Corporation Method of inhibiting gene expression
WO2005003350A2 (fr) 2003-06-27 2005-01-13 Sirna Therapeutics, Inc. Traitement medie par interference d'arn de la maladie d'alzheimer a l'aide d'acide nucleique interferent court (sina)
US20050042647A1 (en) 1996-06-06 2005-02-24 Baker Brenda F. Phosphorous-linked oligomeric compounds and their use in gene modulation
US20050042646A1 (en) 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
GB2415961A (en) 2004-04-14 2006-01-11 Sirna Therapeutics Inc RNA interference (RNAi) of huntingtin (htt) gene
WO2006023544A2 (fr) 2004-08-16 2006-03-02 Quark Biotech, Inc. Applications therapeutiques d'inhibiteurs de rtp801
WO2007084684A2 (fr) 2006-01-20 2007-07-26 Quark Pharmaceuticals, Inc. Utilisations therapeutiques d’inhibiteurs de rtp801
WO2007091269A2 (fr) 2006-02-08 2007-08-16 Quark Pharmaceuticals, Inc. NOVEAU TANDEM d'ARNsi
WO2007107789A2 (fr) 2006-03-17 2007-09-27 Sylentis S.A. Traitement des maladies du snc
WO2007141796A2 (fr) 2006-06-09 2007-12-13 Quark Pharmaceuticals, Inc. Utilisations thérapeutiques d'inhibiteurs de rtp801l
WO2008050329A2 (fr) 2006-10-25 2008-05-02 Quark Pharmaceuticals, Inc. NOUVEAUX ARNsi ET PROCÉDÉS D'UTILISATION DE CEUX-CI
WO2008106102A2 (fr) 2007-02-26 2008-09-04 Quark Pharmaceuticals, Inc. Inhibiteurs de rtp801 et leur utilisation dans le traitement de diverses maladies
US20080287381A1 (en) * 2005-01-30 2008-11-20 Sebastian Thaler Injectable Agent for the Targeted Treatment of Retinal Ganglion Cells
WO2008152636A2 (fr) 2007-06-15 2008-12-18 Quark Pharmaceuticals, Inc. Compositions et procédés d'inhibition de l'expression de la nadph oxydase
WO2009001359A2 (fr) 2007-06-27 2008-12-31 Quark Pharmaceuticals, Inc. Compositions et procédés d'inhibition de l'expression de gènes pro-apoptotiques
WO2009044392A2 (fr) 2007-10-03 2009-04-09 Quark Pharmaceuticals, Inc. Nouvelles structures d'arnsi
WO2009090639A2 (fr) 2008-01-15 2009-07-23 Quark Pharmaceuticals, Inc. Composés d'arnsi et leurs utilisations

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1135105B1 (fr) 1998-12-09 2005-03-02 Chiron Corporation Utilisation d'un agent neurologique pour la preparation d'un medicament pour le traitement d'une maladie du systeme nerveux central
US7825099B2 (en) * 2006-01-20 2010-11-02 Quark Pharmaceuticals, Inc. Treatment or prevention of oto-pathologies by inhibition of pro-apoptotic genes
CN101517081A (zh) * 2006-08-24 2009-08-26 爱尔康研究有限公司 RNAi介导的Gremlin抑制用于治疗眼内压相关的状况
JP5524189B2 (ja) 2008-06-06 2014-06-18 クォーク ファーマシューティカルズ インコーポレーティッド 耳障害治療のための組成物および方法

Patent Citations (80)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3839153A (en) 1970-12-28 1974-10-01 Akzona Inc Process for the detection and determination of specific binding proteins and their corresponding bindable substances
US3791932A (en) 1971-02-10 1974-02-12 Akzona Inc Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3901654A (en) 1971-06-21 1975-08-26 Biological Developments Receptor assays of biologically active compounds employing biologically specific receptors
US3853987A (en) 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
US3867517A (en) 1971-12-21 1975-02-18 Abbott Lab Direct radioimmunoassay for antigens and their antibodies
US3879262A (en) 1972-05-11 1975-04-22 Akzona Inc Detection and determination of haptens
US3850578A (en) 1973-03-12 1974-11-26 H Mcconnell Process for assaying for biologically active molecules
US3935074A (en) 1973-12-17 1976-01-27 Syva Company Antibody steric hindrance immunoassay with two antibodies
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4034074A (en) 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US3984533A (en) 1975-11-13 1976-10-05 General Electric Company Electrophoretic method of detecting antigen-antibody reaction
US4098876A (en) 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
US4879219A (en) 1980-09-19 1989-11-07 General Hospital Corporation Immunoassay utilizing monoclonal high affinity IgM antibodies
US4475196A (en) 1981-03-06 1984-10-02 Zor Clair G Instrument for locating faults in aircraft passenger reading light and attendant call control system
US4447233A (en) 1981-04-10 1984-05-08 Parker-Hannifin Corporation Medication infusion pump
US4439196A (en) 1982-03-18 1984-03-27 Merck & Co., Inc. Osmotic drug delivery system
US4447224A (en) 1982-09-20 1984-05-08 Infusaid Corporation Variable flow implantable infusion apparatus
US4487603A (en) 1982-11-26 1984-12-11 Cordis Corporation Implantable microinfusion pump system
US4486194A (en) 1983-06-08 1984-12-04 James Ferrara Therapeutic device for administering medicaments through the skin
US5011771A (en) 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US4666828A (en) 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4683202B1 (fr) 1985-03-28 1990-11-27 Cetus Corp
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4801531A (en) 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
US4959217A (en) 1986-05-22 1990-09-25 Syntex (U.S.A.) Inc. Delayed/sustained release of macromolecules
US4925678A (en) 1987-04-01 1990-05-15 Ranney David F Endothelial envelopment drug carriers
US5169383A (en) 1988-10-03 1992-12-08 Alza Corporation Control membrane for electrotransport drug delivery
US5272057A (en) 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US6121426A (en) 1988-12-29 2000-09-19 Bio-Technology General Corp. Fibrin binding domain polypeptides and uses and methods of producing same
US5580859A (en) 1989-03-21 1996-12-03 Vical Incorporated Delivery of exogenous DNA sequences in a mammal
US5589466A (en) 1989-03-21 1996-12-31 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5192659A (en) 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5167616A (en) 1989-12-14 1992-12-01 Alza Corporation Iontophoretic delivery method
EP0586520A1 (fr) 1991-05-21 1994-03-16 Isis Pharmaceuticals, Inc. Analogues d'oligonucleotides a squelette modifie
US5225182A (en) 1991-10-31 1993-07-06 Sharma Yash P Vectored drug delivery system using a cephaloplastin linking agent and a methed of using the system
EP0618925B1 (fr) 1991-12-24 2001-08-29 Isis Pharmaceuticals, Inc. Oligonucleotides antisense
WO1993023569A1 (fr) 1992-05-11 1993-11-25 Ribozyme Pharmaceuticals, Inc. Procede et reactif d'inhibition de la replication virale
US5281521A (en) 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
US5593972A (en) 1993-01-26 1997-01-14 The Wistar Institute Genetic immunization
US6586238B1 (en) 1996-04-16 2003-07-01 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids containing 5′-and or 3′-cap structures
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US20050042647A1 (en) 1996-06-06 2005-02-24 Baker Brenda F. Phosphorous-linked oligomeric compounds and their use in gene modulation
US6107094A (en) 1996-06-06 2000-08-22 Isis Pharmaceuticals, Inc. Oligoribonucleotides and ribonucleases for cleaving RNA
WO1998039352A1 (fr) 1997-03-07 1998-09-11 Takeshi Imanishi Nouveaux analogues de bicyclonucleoside et d'oligonucleotide
US6602858B2 (en) 1997-03-31 2003-08-05 Ribozyme Pharmaceuticals, Inc. Nucleic acid catalysts comprising L-nucleotide analogs
WO1999014226A2 (fr) 1997-09-12 1999-03-25 Exiqon A/S Analogues d'oligonucleotides
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
WO2000044914A1 (fr) 1999-01-28 2000-08-03 Medical College Of Georgia Research Institute, Inc. Composition et methode destinees a l'attenuation in vivo et in vitro de l'expression genique utilisant de l'arn double brin
WO2000044895A1 (fr) 1999-01-30 2000-08-03 Roland Kreutzer Methode et medicament destines a inhiber l'expression d'un gene donne
WO2000047599A1 (fr) 1999-02-12 2000-08-17 Sankyo Company, Limited Nouveaux analogues de nucleosides et d'oligonucleotides
WO2000049035A1 (fr) 1999-02-19 2000-08-24 The General Hospital Corporation Inhibition d'un gene
WO2000063364A2 (fr) 1999-04-21 2000-10-26 American Home Products Corporation Procedes et compositions pour l'inhibition de la fonction de sequences polynucleotidiques
WO2001029058A1 (fr) 1999-10-15 2001-04-26 University Of Massachusetts Genes de voies d'interference d'arn en tant qu'outils d'interference genetique ciblee
WO2001036641A2 (fr) 1999-11-02 2001-05-25 Chiron Corporation DETECTEURS DES RECEPTEURS D'ARN DOUBLE BRIN (dsRNA-R) ET PROCEDES CORRESPONDANTS
WO2001036646A1 (fr) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibition d"expression genique a l"aide d"arn bicatenaire
WO2001075164A2 (fr) 2000-03-30 2001-10-11 Whitehead Institute For Biomedical Research Mediateurs d'interference arn specifiques de sequences arn
WO2002044321A2 (fr) 2000-12-01 2002-06-06 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Petites molecules d'arn mediant l'interference arn
WO2002086105A1 (fr) 2001-04-20 2002-10-31 Chiron Corporation Apport d'agents polynucleotides au systeme nerveux central
US20050004064A1 (en) 2001-11-21 2005-01-06 Mitsubishi Chemical Corporation Method of inhibiting gene expression
US20040019001A1 (en) 2002-02-20 2004-01-29 Mcswiggen James A. RNA interference mediated inhibition of protein typrosine phosphatase-1B (PTP-1B) gene expression using short interfering RNA
WO2003087368A2 (fr) * 2002-04-18 2003-10-23 Lynkeus Bio Tech Gmbh Moyens et procedes pour la modulation specifique de genes cibles dans le snc et l'oeil, et procedes pour leur identification
WO2004015107A2 (fr) 2002-08-05 2004-02-19 Atugen Ag Nouvelles formes de molecules d'arn interferant
US7452987B2 (en) 2002-08-05 2008-11-18 Silence Therapeutics Aktiengesellschaft (Ag) Interfering RNA molecules
US20050042646A1 (en) 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
WO2004041924A2 (fr) 2002-11-05 2004-05-21 Isis Pharmaceuticals, Inc. Composes oligomeres a liaison non phosphore et leur utilisation pour la modulation genique
WO2005003350A2 (fr) 2003-06-27 2005-01-13 Sirna Therapeutics, Inc. Traitement medie par interference d'arn de la maladie d'alzheimer a l'aide d'acide nucleique interferent court (sina)
GB2415961A (en) 2004-04-14 2006-01-11 Sirna Therapeutics Inc RNA interference (RNAi) of huntingtin (htt) gene
WO2006023544A2 (fr) 2004-08-16 2006-03-02 Quark Biotech, Inc. Applications therapeutiques d'inhibiteurs de rtp801
US20080287381A1 (en) * 2005-01-30 2008-11-20 Sebastian Thaler Injectable Agent for the Targeted Treatment of Retinal Ganglion Cells
WO2007084684A2 (fr) 2006-01-20 2007-07-26 Quark Pharmaceuticals, Inc. Utilisations therapeutiques d’inhibiteurs de rtp801
WO2007091269A2 (fr) 2006-02-08 2007-08-16 Quark Pharmaceuticals, Inc. NOVEAU TANDEM d'ARNsi
WO2007107789A2 (fr) 2006-03-17 2007-09-27 Sylentis S.A. Traitement des maladies du snc
WO2007141796A2 (fr) 2006-06-09 2007-12-13 Quark Pharmaceuticals, Inc. Utilisations thérapeutiques d'inhibiteurs de rtp801l
WO2008050329A2 (fr) 2006-10-25 2008-05-02 Quark Pharmaceuticals, Inc. NOUVEAUX ARNsi ET PROCÉDÉS D'UTILISATION DE CEUX-CI
WO2008106102A2 (fr) 2007-02-26 2008-09-04 Quark Pharmaceuticals, Inc. Inhibiteurs de rtp801 et leur utilisation dans le traitement de diverses maladies
WO2008152636A2 (fr) 2007-06-15 2008-12-18 Quark Pharmaceuticals, Inc. Compositions et procédés d'inhibition de l'expression de la nadph oxydase
WO2009001359A2 (fr) 2007-06-27 2008-12-31 Quark Pharmaceuticals, Inc. Compositions et procédés d'inhibition de l'expression de gènes pro-apoptotiques
WO2009044392A2 (fr) 2007-10-03 2009-04-09 Quark Pharmaceuticals, Inc. Nouvelles structures d'arnsi
WO2009090639A2 (fr) 2008-01-15 2009-07-23 Quark Pharmaceuticals, Inc. Composés d'arnsi et leurs utilisations

Non-Patent Citations (87)

* Cited by examiner, † Cited by third party
Title
"Comprehensive Medicinal Chemistry", PERGAMON PRESS
"Genome Analysis: A Laboratory Manual Series", vol. 1-4, 1998, COLD SPRING HARBOR LABORATORY PRESS
"Organic syntheses", vol. 1 - 79, 1941, J. WILEY
"PCR Protocols: A Guide To Methods And Applications", 1990, ACADEMIC PRESS
ADAMS ET AL.: "Principles of Neurology", pages: 1057 - 9
ADAMS ET AL.: "Principles of Neurology", pages: 777 - 810
ALLART ET AL., NUCLEOSIDES & NUCLEOTIDES, vol. 17, 1998, pages 1523 - 1526
AMARZGUIOUI ET AL., NAR, vol. 31, no. 2, 2003, pages 589 - 595
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1988, JOHN WILEY AND SONS
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1989, JOHN WILEY AND SONS
BARIK, MOL. MED, vol. 83, 2005, pages 764 - 773
BEAUCAGE; IYER, TETRAHEDRON, vol. 48, 1992, pages 2223 - 2311
BEAUCAGE; IYER, TETRAHEDRON, vol. 49, 1993, pages 6123 - 6194
BEDERSON JB ET AL., STROKE, vol. 17, 1986, pages 472 - 476
BELLON ET AL., BIOCONJUGATE CHEM., vol. 8, 1997, pages 204
BELLON ET AL., NUCLEOSIDES & NUCLEOTIDES, vol. 16, 1997, pages 951
BERNSTEIN ET AL., NATURE, vol. 409, 2001, pages 363 - 6
BERNSTEIN ET AL., NATURE, vol. 409, no. 6818, 2001, pages 363 - 6
BERNSTEIN ET AL., RNA, vol. 7, no. 11, 2001, pages 1509 - 21
BITKO ET AL.: "showed that specific siRNAs directed against the respiratory syncytial virus (RSV) nucleocapsid N gene are effective in treating mice when administered intranasally", NAT. MED., vol. 11, no. 1, 2005, pages 50 - 55
BRAASCH ET AL., BIOCHEM., vol. 42, no. 26, 2003, pages 7967 - 75
BRAIN PATHOL, vol. 8, no. 2, April 1998 (1998-04-01), pages 339 - 54
CARUTHERS, METHODS ENZYMOL., vol. 154, 1987, pages 287 - 313
CHAKRABORTY, CURRENT DRUG TARGETS, vol. 8, no. 3, 2007, pages 469 - 82
CHALK ET AL., BBRC., vol. 319, no. 1, 2004, pages 264 - 74
CHEN Y ET AL., J. NEUROTRAUMA., vol. 13, 1996, pages 557 - 568
CHIU ET AL., RNA, vol. 9, no. 9, 2003, pages 1034 - 48
CHIU; RANA, RNA, vol. 9, 2003, pages 1034 - 1048
CZAUDERNA ET AL., NAR, vol. 31, no. 11, 2003, pages 2705 - 2716
DOGAN A. ET AL., J. NEUROCHEM., vol. 72, 1999, pages 765 - 770
DURCAN, MOL. PHARMA., vol. 5, no. 4, 2008, pages 559 - 566
ECKSTEIN, ANN. REV. BIOCHEM., vol. 54, 1985, pages 367 - 402
ELBASHIR ET AL., GENES DEV., vol. 15, 2001, pages 188
ELBASHIR ET AL., GENES DEV., vol. 15, no. 2, 2001, pages 188 - 200
ELMEN ET AL., NAR, vol. 33, no. 1, 2005, pages 439 - 447
FISHER ET AL., NAR, vol. 35, no. 4, 2007, pages 1064 - 1074
GEWERT ET AL.: "Organic synthesis workbook", 2000, WILEY-VCH
GRIMM; KAY, JCI, vol. 117, no. 12, 2007, pages 3633 - 41
GRUNER JA.: "A monitored contusion model of spinal cord injury in the rat.", J. NEUROTRAUMA, vol. 9, no. 2, 1992, pages 123 - 128
HASEGAWA K; GRUMET M.: "Trauma-induced tumorigenesis of cells implanted into the rat spinal cord.", J. NEUROSURG., vol. 98, no. 5, 2003, pages 1065 - 71
HERDEWIJN ET AL., NUCLEOSIDES & NUCLEOTIDES, vol. 18, 1999, pages 1371 - 1376
HOLEN ET AL., NAR, vol. 31, no. 9, 2003, pages 2401 - 2407
HUANG PP; YOUNG W.: "The effects of arterial blood gas values on lesion volumes in a graded rat spinal cord contusion model.", J NEUROTRAUMA, vol. 11, no. 5, 1994, pages 547 - 562
KAISER, AM J OPHTHALMOL., vol. 142, no. 4, 2006, pages 660 - 8
KIERAN D ET AL., PNAS, vol. 104, no. 51, 2007, pages 20606 - 20611
KIERAN ET AL., J CELL BIOL., vol. 169, 2005, pages 561 - 567
KIM; ROSSI, BIOTECHNIQUES, vol. 44, 2008, pages 613 - 616
KNIGHT RA ET AL., STROKE, vol. 25, 1994, pages 1252 - 1261
LEE ET AL., NATURE, vol. 425, 2003, pages 415 - 9
LEE ET AL., NATURE, vol. 425, no. 6956, 2003, pages 415 - 419
LEVENKOVA ET AL., BIOINFORM., vol. 20, no. 3, 2004, pages 430 - 2
LONGA EZ ET AL., STROKE, vol. 20, 1989, pages 84 - 91
MAEDA YUKIHIDE ET AL: "Therapeutic regulation of gene expression in the inner ear using RNA interference.", NIH PUBLIC ACCESS AUTHOR MANUSCRIPT, 11 May 2010 (2010-05-11), pages 1 - 21, XP002631052, Retrieved from the Internet <URL:http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2867253/pdf/nihms183708.pdf?tool=pmcentrez> [retrieved on 20110401] *
MCMANUS; SHARP, NATURE REV GENET, vol. 3, no. 10, 2002, pages 737 - 47
MINTOROVITCH J. ET AL., MAGN. RESON. MED., vol. 18, 1991, pages 39 - 50
MOECHARS D. ET AL., EMBO J., vol. 15, no. 6, 1996, pages 1265 - 74
MOECHARS D. ET AL., NEUROSCIENCE, vol. 91, no. 3, 1999, pages 819 - 30
MONTOYA CP ET AL., J. NEUROSCI. METHODS, vol. 36, 1991, pages 219 - 228
MOORE ET AL., SCIENCE, vol. 256, 1992, pages 9923
NISHIKURA, CELL, vol. 107, no. 4, 2001, pages 415 - 8
PADDISON; HANNON, CURR OPIN MOL THER., vol. 5, no. 3, 2003, pages 217 - 24
PEASE ET AL., J. GLAUCOMA, vol. 15, no. 6, 2006, pages 512 - 9
PERBAL: "A Practical Guide to Molecular Cloning", 1988, JOHN WILEY & SONS
RAOUL C ET AL., NATURE MED., vol. 11, 2005, pages 423 - 428
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRINGS HARBOR
SAMBROOK ET AL.: "Molecular cloning: A laboratory manual", 1989, COLD SPRINGS HARBOR LABORATORY
SCARINGE ET AL., NAR, vol. 18, 1990, pages 5433
SHABAROVA ET AL., NAR, vol. 19, 1991, pages 4247
SHARP, NATURE REV GENET, vol. 3, 2002, pages 737 - 47
SIOUD; LEIRDAL, MET. MOL BIOL., vol. 252, 2004, pages 457 - 69
SMITH; MARCH: "Advanced Organic Chemistry", 2001, WILEY-INTERSCIENCE
SONG ET AL.: "Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors", NAT BIOTECHNOL., vol. 23, no. 6, 2005, pages 709 - 17, XP002390626, DOI: doi:10.1038/nbt1101
TAKEI ET AL., JBC, vol. 277, no. 26, 2002, pages 23800 - 06
TAMURA A. ET AL., J CEREB BLOOD FLOW METAB., vol. 1, 1981, pages 53 - 60
TESTONI ET AL., BLOOD, vol. 87, 1996, pages 3822
TOWNE C ET AL., MOL THER., vol. 16, 2008, pages 1018 - 1025
UI-TEI ET AL., J BIOMED BIOTECHNOL., vol. 2006, 2006, pages 65052
UI-TEI ET AL., J BIOMED BIOTECHNOL., vol. 65052, 2006
UI-TEI ET AL., NAR, vol. 32, no. 3, 2004, pages 936 - 48
UI-TEI ET AL., NUC. ACID RES., vol. 32, no. 3, 2004, pages 936 - 48
UI-TEI, NAR, vol. 36, no. 7, 2008, pages 2136 - 2151
USMAN ET AL., J. AM. CHEM. SOC., vol. 109, 1987, pages 7845
WATSON ET AL.: "Scientific American Books", article "Recombinant DNA"
WINCOTT ET AL., METHODS MOL. BIO., vol. 74, 1997, pages 59
WINCOTT ET AL., NAR, vol. 23, 1995, pages 2677 - 2684
YOUNG, PROG BRAIN RES., vol. 137, 2002, pages 231 - 55
YU-LAI WANG ET AL.: "Clinico-pathological rescue of a model mouse of Huntington's disease by siRNA", NEUROSCI RES, vol. 53, no. 3, pages 241 - 9

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9828420B2 (en) 2007-01-05 2017-11-28 University Of Zürich Method of providing disease-specific binding molecules and targets
US10131708B2 (en) 2007-01-05 2018-11-20 University Of Zürich Methods of treating Alzheimer's disease
WO2013037499A1 (fr) * 2011-09-15 2013-03-21 Johann Wolfgang Goethe-Universität Inhibiteurs de l'expression de nox4 et/ou de la fonction de nox4 et leur utilisation pour la prévention et le traitement des lésions nerveuses et/ou de la douleur neuropathique
WO2013067076A3 (fr) * 2011-11-03 2013-07-04 Quark Pharmaceuticals, Inc. Procédés et compositions pour la neuroprotection
CN103917647B (zh) * 2011-11-03 2020-07-10 夸克制药公司 用于神经保护的方法和组合物
CN103917647A (zh) * 2011-11-03 2014-07-09 夸克制药公司 用于神经保护的方法和组合物
KR20140091683A (ko) * 2011-11-03 2014-07-22 쿠아크 파마수티칼스 인코퍼레이티드 신경보호를 위한 방법 및 조성물
AU2012332517B9 (en) * 2011-11-03 2017-08-10 Quark Pharmaceuticals, Inc. Methods and compositions for neuroprotection
KR101970034B1 (ko) 2011-11-03 2019-04-17 쿠아크 파마수티칼스 인코퍼레이티드 신경보호를 위한 방법 및 조성물
WO2013067076A2 (fr) 2011-11-03 2013-05-10 Quark Pharmaceuticals, Inc. Procédés et compositions pour la neuroprotection
US9422560B2 (en) 2011-11-03 2016-08-23 Quark Pharmaceuticals, Inc. Methods and compositions for neuroprotection
US9738896B2 (en) 2011-11-03 2017-08-22 Quark Pharmaceuticals, Inc. Methods and compositions for neuroprotection
WO2013070821A1 (fr) 2011-11-08 2013-05-16 Quark Pharmaceuticals, Inc. Méthodes et compositions destinées à traiter des maladies, des troubles ou une lésion du système nerveux
US10570394B2 (en) 2012-01-04 2020-02-25 Quark Pharmaceuticals, Inc. Double-stranded RNA compounds to CASP2 and uses thereof
EA036003B1 (ru) * 2012-01-04 2020-09-11 Кварк Фармасьютикалс, Инк. Способ лечения оптической нейропатии с применением двухцепочечной рнк к casp2
US9382542B2 (en) 2012-01-04 2016-07-05 Quark Pharmaceuticals, Inc. Double-stranded RNA compounds to CASP2 and uses thereof
KR102202004B1 (ko) 2012-01-04 2021-01-12 쿠아크 파마수티칼스 인코퍼레이티드 Casp2에 대한 이중-가닥 rna 화합물 및 그 용도
KR20190137940A (ko) * 2012-01-04 2019-12-11 쿠아크 파마수티칼스 인코퍼레이티드 Casp2에 대한 이중-가닥 rna 화합물 및 그 용도
WO2013103632A1 (fr) * 2012-01-04 2013-07-11 Quark Pharmaceuticals, Inc. Composés arn double brin dirigés contre casp2 et leurs utilisations
WO2015006475A1 (fr) * 2013-07-12 2015-01-15 Biogen Idec International Neuroscience Gmbh Biomarqueurs génétiques et d'imagerie associés au déclin des mesures cognitives et du métabolisme cérébral du glucose chez des sujets souffrant de la maladie d'alzheimer ou risquant de la développer
US11401516B2 (en) 2014-08-22 2022-08-02 Auckland Uniservices Limited Channel modulators
WO2016029191A3 (fr) * 2014-08-22 2016-08-18 Auckland Uniservices Limited Modulateurs de canaux
US10465188B2 (en) 2014-08-22 2019-11-05 Auckland Uniservices Limited Channel modulators
CN104357470A (zh) * 2014-11-14 2015-02-18 四川大学华西第二医院 一种HtrA2融合基因及其应用
CN104357470B (zh) * 2014-11-14 2017-02-15 四川大学华西第二医院 一种HtrA2融合基因及其应用
US10842871B2 (en) 2014-12-02 2020-11-24 Biogen International Neuroscience Gmbh Methods for treating Alzheimer's disease
EP4035659A1 (fr) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes destinés à l'administration d'agents thérapeutiques
CN106967789A (zh) * 2017-01-26 2017-07-21 上海长海医院 一种前列腺癌标志物plxna1及其应用
US11655289B2 (en) 2017-08-22 2023-05-23 Biogen Ma Inc. Pharmaceutical compositions containing anti-beta amyloid antibodies
CN112716971A (zh) * 2021-01-19 2021-04-30 苏州大学 lncRNA XR_595534.2在制备治疗或预防慢性疼痛的药物中的应用

Also Published As

Publication number Publication date
EP2862929B1 (fr) 2017-09-06
US20120252875A1 (en) 2012-10-04
EP2510098B1 (fr) 2015-02-11
EP2862929A1 (fr) 2015-04-22
US8778904B2 (en) 2014-07-15
EP2510098A1 (fr) 2012-10-17

Similar Documents

Publication Publication Date Title
EP2510098B1 (fr) Méthodes et compositions utilisées pour le traitement de maladies, d&#39;affections ou de lésions du snc
US10421962B2 (en) Double-stranded oligonucleotide molecules to DDIT4 and methods of use thereof
JP5592892B2 (ja) 眼障害の治療方法
CA2577423C (fr) Applications therapeutiques d&#39;inhibiteurs de rtp801
RU2426544C2 (ru) Лечение нарушений цнс
US9056903B2 (en) Therapeutic uses of inhibitors of RTP801
WO2007141796A2 (fr) Utilisations thérapeutiques d&#39;inhibiteurs de rtp801l
US20110034534A1 (en) siRNA compounds and methods of use thereof
WO2011057171A1 (fr) Méthodes d&#39;administration d&#39;arnsi dans la moelle épinière, et thérapies qui en découlent
US20110028531A1 (en) Novel sirna compounds for inhibiting rtp801
WO2010080452A2 (fr) Composés d&#39;arnsi et leurs procédés d&#39;utilisation
US20140350068A1 (en) Rtp801l sirna compounds and methods of use thereof
SG186438A1 (en) Double stranded rna compounds to rhoa and use thereof
WO2009074990A2 (fr) Composes de petits arn interferents ciblant rtp801l et methodes d&#39;utilisation associees
US20140323549A1 (en) Methods and compositions for treating diseases, disorders or injury of the nervous system

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: 10809113

Country of ref document: EP

Kind code of ref document: A1

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

Ref document number: 10809113

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 13514880

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2010809113

Country of ref document: EP