US20200399641A1 - Antisense oligonucleotides for modulating htra1 expression - Google Patents

Antisense oligonucleotides for modulating htra1 expression Download PDF

Info

Publication number
US20200399641A1
US20200399641A1 US16/730,779 US201916730779A US2020399641A1 US 20200399641 A1 US20200399641 A1 US 20200399641A1 US 201916730779 A US201916730779 A US 201916730779A US 2020399641 A1 US2020399641 A1 US 2020399641A1
Authority
US
United States
Prior art keywords
oligonucleotide
htra1
nucleosides
seq
lna
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/730,779
Inventor
Roberto Iacone
Peter Hagedorn
Susanne Kammler
Soren Ottosen
Sindri Traustason
Heidi Hudlebusch
Lykke Pedersen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Hoffmann La Roche Inc
Original Assignee
Hoffmann La Roche 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 Hoffmann La Roche Inc filed Critical Hoffmann La Roche Inc
Priority to US16/730,779 priority Critical patent/US20200399641A1/en
Publication of US20200399641A1 publication Critical patent/US20200399641A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified C
    • C12N2310/33415-Methylcytosine
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/341Gapmers, i.e. of the type ===---===
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/344Position-specific modifications, e.g. on every purine, at the 3'-end
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/346Spatial arrangement of the modifications having a combination of backbone and sugar modifications
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate

Definitions

  • the present invention relates to antisense oligonucleotides (oligomers) that are complementary to HTRA1, leading to modulation of the expression of HTRA1. Modulation of HTRA1 expression is beneficial for a range of medical disorders, such as macular degeneration, e.g. age-related macular degeneration.
  • HTRA human high temperature requirement A
  • HTRA housekeeping proteases are implicated in organization of the extracellular matrix, cell proliferation and ageing. Modulation of HTRA activity is connected with severe diseases, including Duchenne muscular dystrophy (Bakay et al. 2002, Neuromuscul. Disord. 12: 125-141), arthritis, such as osteoarthritis (Grau et al.
  • HTRA1 insulin-like growth factor (IGF) binding domain. It has been proposed to regulate IGF availability and cell growth (Zumbrunn and Trueb, 1996, FEES Letters 398:189-192) and to exhibit tumor suppressor properties. HTRA1 expression is down-regulated in metastatic melanoma, and may thus indicate the degree of melanoma progression.
  • IGF insulin-like growth factor
  • HTRA1 expression is also down-regulated in ovarian cancer. In ovarian cancer cell lines, HTRA1 overexpression induces cell death, while antisense HTRA1 expression promoted anchorage-independent growth (Chien et al., 2004, Oncogene 23:1636-1644).
  • HTRA1 In addition to its effect on the IGF pathway, HTRA1 also inhibits signaling by the TGF ⁇ family of growth factors (Oka et al., 2004, Development 131:1041-1053). HTRA1 can cleave amyloid precursor protein (APP), and HTRA1 inhibitors cause the accumulation of A ⁇ peptide in cultured cells. Thus, HTRA1 is also implicated in Alzheimer's disease (Grau et al., 2005, Proc. Nat. Acad. Sci. USA. 102:6021-6026).
  • APP amyloid precursor protein
  • HTRA1 upregulation has been observed and seems to be associated to Duchenne muscular dystrophy (Bakay et al. 2002, Neuromuscul. Disord. 12: 125-141) and osteoarthritis (Grau et al. 2006, JBC 281: 6124-6129) and AMD (Fritsche, et al. Nat Gen 2013 45(4):433-9.)
  • a single nucleotide polymorphism (SNP) in the HTRA1 promoter region is associated with a 10 fold increased the risk of developing age-related macular degeneration (AMD).
  • the HTRA1 SNPs are in linkage disequilibrium with the ARMS2 SNP (rs10490924) associated with increased risk of developing age-related macular degeneration (AMD).
  • the risk allele is associated with 2-3 fold increased HTRA1 mRNA and protein expression, and HTRA1 is present in drusen in patients with AMD (Dewan et al., 2006, Science 314:989-992; Yang et al., 2006, Science 314:992-993).
  • HtrA1 Induces AMD-like phenotype in mice.
  • the hHTRA transgenic mouse (Veierkottn, PlosOne 2011) reveals degradation of the elastic lamina of Bruch's membrane, determines choroidal vascular abnormalities (Jones, PNAS 2011) and increases the Polypoidal choroidal vasculopathy (PCV) lesions (Kumar, IOVS 2014). Additionally it has been reported Bruch's membrane damage in hHTRA1 Tg mice, which determines upon exposure to cigarette smoke 3 fold increases CNV (Nakayama, IOVS 2014)
  • Age-related macular degeneration is the leading cause of irreversible loss of vision in people over the age of 65. With onset of AMD there is gradual loss of the light sensitive photoreceptor cells in the back of the eye, the underlying pigment epithelial cells that support them metabolically, and the sharp central vision they provide. Age is the major risk factor for the onset of AMD: the likelihood of developing AMD triples after age 55. Smoking, light iris color, gender (women are at greater risk), obesity, and repeated exposure to UV radiation also increase the risk of AMD. There are two forms of AMD: dry AMD and wet AMD. In dry AMD, drusen appear in the macula of the eye, the cells in the macula die, and vision becomes blurry.
  • Dry AMD can progress in three stages: 1) early, 2) intermediate, and 3) advanced dry AMD. Dry AMD can also progress into wet AMD during any of these stages.
  • Wet AMD also known as exudative AMD
  • PSNV posterior segment neovascularization
  • the posterior segment neovascularization (PSNV) found in exudative AMD is characterized as pathologic choroidal neovascularization. Leakage from abnormal blood vessels forming in this process damages the macula and impairs vision, eventually leading to blindness. Treatment strategies for wet AMD are few and palliative at best. There is therefore an unmet medical need in the provision of effective drugs to treat macular degenerative conditions such as wet and dry AMD.
  • WO 2008/013893 claims a composition for treating a subject suffering from age related macular degeneration comprising a nucleic acid molecules comprising an antisense sequence that hybridizes to HTRA1 gene or mRNA: No antisense molecules are disclosed.
  • WO2009/006460 provides siRNAs targeting HTRA1 and their use in treating AMD.
  • the present invention provides antisense oligonucleotides which modulate HTRA1 in vivo or in vitro.
  • the invention identified cryptic target sequence motifs present in the human HTRA1 mRNA (including pre-mRNA) which may be targeted by antisense oligonucleotides to give effective HTRA1 inhibition.
  • the invention also provides effective antisense oligonucleotide sequences and compounds which are capable of inhibiting HTRA1, and their use in treatment of diseases or disorders where HTRA1 is indicated.
  • the present invention relates to oligonucleotides targeting a mammalian HTRA1 nucleic acid, i.e. are capable of inhibiting the expression of HTRA1 and to treat or prevent diseases related to the functioning of the HTRA1.
  • the oligonucleotides targeting HTRA1 are antisense oligonucleotides, i.e. are complementary to their HTRA1 nucleic acid target.
  • the oligonucleotide of the invention may be in the form of a pharmaceutically acceptable salt, such as a sodium salt or a potassium salt.
  • the invention provides antisense oligonucleotides which comprise a contiguous nucleotide sequence of 10-30 nucleotides in length with at least 90% complementarity, such as fully complementary to a mammalian HTRA1 nucleic acid, such as SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 3 or SEQ ID NO 4.
  • the invention provides pharmaceutical compositions comprising the oligonucleotides of the invention and pharmaceutically acceptable diluents, carriers, salts and/or adjuvants.
  • LNA antisense oligonucleotides such as LNA gapmer oligonucleotides, which comprise a contiguous nucleotide sequence of 10-30 nucleotides in length with at least 90% complementarity, such as fully complementary to a HTRA1 nucleic acid, such as a sequence selected from the group consisting of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 3 or SEQ ID NO 4.
  • the invention provides for an antisense oligonucleotide comprising a contiguous nucleotide region of 10-22, such as 12-22 nucleotides which are at least 90% such as 100% complementarity to SEQ ID NO 147:
  • the invention provides for an antisense oligonucleotide comprising a contiguous nucleotide region of 10-17, such as 11, 12, 13, 14, 15, 16, such as 12-16 or 12-17 nucleotides which are complementarity to a sequence selected from the group consisting of SEQ ID NO 148-155.
  • the invention provides for an antisense oligonucleotide comprising a contiguous nucleotide region of 10-17, such as 11, 12, 13, 14, 15, 16, such as 12-16 or 12-17 nucleotides which are complementarity to SEQ ID NO 148 or 155.
  • the invention provides for an antisense oligonucleotide of 10-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of 10-22 nucleotides which are at least 90% such as 100% complementarity to SEQ ID NO 147:
  • the invention provides for an antisense oligonucleotide of 10-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of at least 10, such as at least 12 contiguous nucleotides which are complementary to a sequence present in a sequence selected from SEQ ID NO 148-155.
  • the invention provides for an antisense oligonucleotide of at least 12 nucleotides in length, wherein said antisense oligonucleotide comprises the contiguous sequence of SEQ ID NO 146
  • SEQ ID NO 146 5′ TTTACCTGGTT 3′.
  • the invention provides for the oligonucleotides provided in the examples.
  • the invention provides for the oligonucleotide, such as an antisense oligonucleotide, which comprises at least 10, such as at least 12, present in a sequence selected from the group consisting of SEQ ID NO 5-145.
  • the invention provides for a conjugate comprising the oligonucleotide according to the invention, and at least one conjugate moiety covalently attached to said oligonucleotide.
  • the invention provides for a pharmaceutically acceptable salt of the oligonucleotide or conjugate of the invention.
  • the invention provides methods for in vivo or in vitro method for modulation of HTRA1 expression in a cell which is expressing HTRA1, by administering an oligonucleotide, conjugate or composition of the invention in an effective amount to said cell.
  • the invention provides methods for treating or preventing a disease, disorder or dysfunction associated with in vivo activity of HTRA1 comprising administering a therapeutically or prophylactically effective amount of the oligonucleotide of the invention, or conjugate thereof, to a subject suffering from or susceptible to the disease, disorder or dysfunction.
  • oligonucleotide or composition of the invention is used for the treatment or prevention of macular degeneration, and other disorders where HTRA1 is implicated.
  • the invention provides for the oligonucleotide or conjugate of the invention, for use in the treatment of a disease or disorder selected from the list comprising of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, Alzhiemer's disease and Parkinson's disease.
  • a disease or disorder selected from the list comprising of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, Alzhiemer's disease and Parkinson's disease.
  • the invention provides for the oligonucleotide or conjugate of the invention, for use in the treatment of macular degeneration, such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy.
  • macular degeneration such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy.
  • the invention provides for the use of the oligonucleotide, conjugate or composition of the invention, for the manufacture of a medicament for the treatment of macular degeneration, such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy.
  • macular degeneration such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy.
  • the invention provides for the use of the oligonucleotide, conjugate or composition of the invention, for the manufacture of a medicament for the treatment of a disease or disorder selected from the group consisting of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, Alzhiemer's disease and Parkinson's disease.
  • a disease or disorder selected from the group consisting of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, Alzhiemer's disease and Parkinson's disease.
  • the invention provides for a method of treatment of a subject suffering from a disease or disorder selected from the group consisting of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, Alzhiemer's disease and Parkinson's disease, said method comprising the step of administering an effective amount of the oligonucleotide, conjugate or composition of the invention to the subject.
  • a disease or disorder selected from the group consisting of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, Alzhiemer's disease and Parkinson's disease
  • the invention provides for a method of treatment of a subject suffering from an ocular disease, such as macular degeneration, such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy, said method comprising the step of administering an effective amount of the oligonucleotide, conjugate or composition of the invention to the subject.
  • macular degeneration such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy
  • the invention provides for a method of treatment of a subject suffering from an ocular disease, such as macular degeneration, such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy, said method comprising administering at least two dosages of the oligonucleotide of the invention, or pharmaceutically acceptable salt thereof, in an intraocular injection in a dosage of from about 10 ⁇ g-200 ⁇ g, wherein the dosage interval between administration consecutive is at least 4 weeks or at least monthly.
  • macular disease such as macular degeneration, such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy
  • FIG. 3 Dose response of HTRA1 mRNA level upon treatment of human primary RPE cells with LNA oligonucleotide 139.1 and 143.1.
  • FIG. 4 Results of analysis of in vivo expression of HTRA1.
  • FIGS. 5A-5D Rat in vivo efficacy study, 7 days of treatment, IVT administration, dose response.
  • Retina samples from rat eyes treated with PBS, 140.1 or 143.1 were analyzed.
  • Htra1 ISH RNAscope was performed, in A) representative samples and in B) an overview table of results are shown.
  • GFAP IHC was also performed, GFAP is a marker for reactive gliosis and reticular fibrosis.
  • FIGS. 6A-6D Poc study, Blue light-induced retinal degeneration in albino rats
  • FIGS. 7A-7C Rat in vivo efficacy kinetic study, IVT administration, 50 ⁇ g/eye, 3, 7, 14 days of treatment.
  • FIGS. 8A-8D Non Human Primate (NHP) PK/PD study, IVT administration, 25 ⁇ g/eye.
  • C-D) Quantification of HTRA1 protein level in retina and vitreous, respectively, by IP-MS. Dots show data for individual animals. Error bars show standard deviations for technical replicates (n 3).
  • FIG. 9 A Compound of the invention (Compound ID NO 143.1).
  • the compound may be in the form of a pharmaceutical salt, such as a sodium salt or a potassium salt.
  • FIG. 10 A Compound of the invention (Compound ID No 145.3).
  • the compound may be in the form of a pharmaceutical salt, such as a sodium salt or a potassium salt.
  • FIG. 11 An example of a pharmaceutical salt of compound 143.1.
  • M+ is a suitable cation, typically a positive metal ion, such as a sodium or potassium ion.
  • the stoichiometric ratio of the cation to the oligonucleotide anion will depend on the charge of the cation used.
  • cations with one, two or three positive charge may be used.
  • twice as many single+charged cations (monovalent), such as Na + or K + are needed as compared to a divalent cation such as Ca 2+ .
  • FIG. 12 An example of a pharmaceutical salt of compound 145.3. See the figure legend for FIG. 11 for the description of the cation M.
  • oligonucleotide as used herein is defined as it is generally understood by the skilled person as a molecule comprising two or more covalently linked nucleosides. Such covalently bound nucleosides may also be referred to as nucleic acid molecules or oligomers. Oligonucleotides are commonly made in the laboratory by solid-phase chemical synthesis followed by purification. When referring to a sequence of the oligonucleotide, reference is made to the sequence or order of nucleobase moieties, or modifications thereof, of the covalently linked nucleotides or nucleosides.
  • the oligonucleotide of the invention is man-made, and is chemically synthesized, and is typically purified or isolated.
  • the oligonucleotide of the invention may comprise one or more modified nucleosides or nucleotides.
  • Antisense oligonucleotide as used herein is defined as oligonucleotides capable of modulating expression of a target gene by hybridizing to a target nucleic acid, in particular to a contiguous sequence on a target nucleic acid.
  • the antisense oligonucleotides are not essentially double stranded and are therefore not siRNAs.
  • the antisense oligonucleotides of the present invention are single stranded.
  • contiguous nucleotide region refers to the region of the oligonucleotide which is complementary to the target nucleic acid.
  • the term may be used interchangeably herein with the term “contiguous nucleotide sequence” or “contiguous nucleobase sequence” and the term “oligonucleotide motif sequence”. In some embodiments all the nucleotides of the oligonucleotide are present in the contiguous nucleotide region.
  • the oligonucleotide comprises the contiguous nucleotide region and may, optionally comprise further nucleotide(s), for example a nucleotide linker region which may be used to attach a functional group to the contiguous nucleotide sequence.
  • the nucleotide linker region may or may not be complementary to the target nucleic acid.
  • the internucleoside linkages present between the nucleotides of the contiguous nucleotide region are all phosphorothioate internucleoside linkages.
  • the contiguous nucleotide region comprises one or more sugar modified nucleosides.
  • Nucleotides are the building blocks of oligonucleotides and polynucleotides, and for the purposes of the present invention include both naturally occurring and non-naturally occurring nucleotides.
  • nucleotides such as DNA and RNA nucleotides comprise a ribose sugar moiety, a nucleobase moiety and one or more phosphate groups (which is absent in nucleosides).
  • Nucleosides and nucleotides may also interchangeably be referred to as “units” or “monomers”.
  • modified nucleoside or “nucleoside modification” as used herein refers to nucleosides modified as compared to the equivalent DNA or RNA nucleoside by the introduction of one or more modifications of the sugar moiety or the (nucleo)base moiety.
  • the modified nucleoside comprise a modified sugar moiety.
  • modified nucleoside may also be used herein interchangeably with the term “nucleoside analogue” or modified “units” or modified “monomers”.
  • modified internucleoside linkage is defined as generally understood by the skilled person as linkages other than phosphodiester (PO) linkages, that covalently couples two nucleosides together. Nucleotides with modified internucleoside linkage are also termed “modified nucleotides”. In some embodiments, the modified internucleoside linkage increases the nuclease resistance of the oligonucleotide compared to a phosphodiester linkage. For naturally occurring oligonucleotides, the internucleoside linkage includes phosphate groups creating a phosphodiester bond between adjacent nucleosides.
  • Modified internucleoside linkages are particularly useful in stabilizing oligonucleotides for in vivo use, and may serve to protect against nuclease cleavage at regions of DNA or RNA nucleosides in the oligonucleotide of the invention, for example within the gap region of a gapmer oligonucleotide, as well as in regions of modified nucleosides.
  • the oligonucleotide comprises one or more internucleoside linkages modified from the natural phosphodiester to a linkage that is for example more resistant to nuclease attack.
  • Nuclease resistance may be determined by incubating the oligonucleotide in blood serum or by using a nuclease resistance assay (e.g. snake venom phosphodiesterase (SVPD)), both are well known in the art.
  • SVPD snake venom phosphodiesterase
  • Internucleoside linkages which are capable of enhancing the nuclease resistance of an oligonucleotide are referred to as nuclease resistant internucleoside linkages.
  • all of the internucleoside linkages of the oligonucleotide, or contiguous nucleotide sequence thereof are modified. It will be recognized that, in some embodiments the nucleosides which link the oligonucleotide of the invention to a non-nucleotide functional group, such as a conjugate, may be phosphodiester. In some embodiments all of the internucleoside linkages of the oligonucleotide, or contiguous nucleotide sequence thereof, are nuclease resistant internucleoside linkages.
  • the modified internucleoside linkages may be phosphorothioate internucleoside linkages. In some embodiments, the modified internucleoside linkages are compatible with the RNaseH recruitment of the oligonucleotide of the invention, for example phosphorothioate.
  • the internucleoside linkage comprises sulphur (S), such as a phosphorothioate internucleoside linkage.
  • a phosphorothioate internucleoside linkage is particularly useful due to nuclease resistance, beneficial pharmacokinetics and ease of manufacture.
  • all of the internucleoside linkages of the oligonucleotide, or contiguous nucleotide sequence thereof, are phosphorothioate.
  • nucleobase includes the purine (e.g. adenine and guanine) and pyrimidine (e.g. uracil, thymine and cytosine) moiety present in nucleosides and nucleotides which form hydrogen bonds in nucleic acid hybridization.
  • pyrimidine e.g. uracil, thymine and cytosine
  • nucleobase also encompasses modified nucleobases which may differ from naturally occurring nucleobases, but are functional during nucleic acid hybridization.
  • nucleobase refers to both naturally occurring nucleobases such as adenine, guanine, cytosine, thymidine, uracil, xanthine and hypoxanthine, as well as non-naturally occurring variants. Such variants are for example described in Hirao et al (2012) Accounts of Chemical Research vol 45 page 2055 and Bergstrom (2009) Current Protocols in Nucleic Acid Chemistry Suppl. 37 1.4.1.
  • the nucleobase moiety is modified by changing the purine or pyrimidine into a modified purine or pyrimidine, such as substituted purine or substituted pyrimidine, such as a nucleobased selected from isocytosine, pseudoisocytosine, 5-methyl cytosine, 5-thiozolo-cytosine, 5-propynyl-cytosine, 5-propynyl-uracil, 5-bromouracil 5-thiazolo-uracil, 2-thio-uracil, 2′thio-thymine, inosine, diaminopurine, 6-aminopurine, 2-aminopurine, 2,6-diaminopurine and 2-chloro-6-aminopurine.
  • a nucleobased selected from isocytosine, pseudoisocytosine, 5-methyl cytosine, 5-thiozolo-cytosine, 5-propynyl-cytosine, 5-propynyl-uracil, 5-brom
  • the nucleobase moieties may be indicated by the letter code for each corresponding nucleobase, e.g. A, T, G, C or U, wherein each letter may optionally include modified nucleobases of equivalent function.
  • the nucleobase moieties are selected from A, T, G, C, and 5-methyl cytosine.
  • 5-methyl cytosine LNA nucleosides may be used.
  • the cytosine nucleobases in a 5′cg3′ motif is 5-methyl cytosine.
  • modified oligonucleotide describes an oligonucleotide comprising one or more sugar-modified nucleosides and/or modified internucleoside linkages.
  • chimeric oligonucleotide is a term that has been used in the literature to describe oligonucleotides with modified nucleosides.
  • complementarity describes the capacity for Watson-Crick base-pairing of nucleosides/nucleotides.
  • Watson-Crick base pairs are guanine (G)—cytosine (C) and adenine (A)—thymine (T)/uracil (U).
  • G guanine
  • A cytosine
  • T thymine
  • U uracil
  • oligonucleotides may comprise nucleosides with modified nucleobases, for example 5-methyl cytosine is often used in place of cytosine, and as such the term complementarity encompasses Watson Crick base-paring between non-modified and modified nucleobases (see for example Hirao et al (2012) Accounts of Chemical Research vol 45 page 2055 and Bergstrom (2009) Current Protocols in Nucleic Acid Chemistry Suppl. 37 1.4.1).
  • % complementary refers to the number of nucleotides in percent of a contiguous nucleotide region or sequence in a nucleic acid molecule (e.g. oligonucleotide) which, at a given position, are complementary to (i.e. form Watson Crick base pairs with) a contiguous nucleotide sequence, at a given position of a separate nucleic acid molecule (e.g. the target nucleic acid).
  • the percentage is calculated by counting the number of aligned bases that form pairs between the two sequences, dividing by the total number of nucleotides in the oligonucleotide and multiplying by 100. In such a comparison a nucleobase/nucleotide which does not align (form a base pair) is termed a mismatch.
  • complementarity when referring to complementarity between two sequences, the determination of complementarity is measured across the length of the shorter of the two sequences, such as the length of the contiguous nucleotide region or sequence.
  • Identity refers to the number of nucleotides in percent of a contiguous nucleotide sequence in a nucleic acid molecule (e.g. oligonucleotide) which, at a given position, are identical to (i.e. in their ability to form Watson Crick base pairs with the complementary nucleoside) a contiguous nucleotide sequence, at a given position of a separate nucleic acid molecule (e.g. the target nucleic acid).
  • the percentage is calculated by counting the number of aligned bases that are identical between the two sequences, including gaps, dividing by the total number of nucleotides in the oligonucleotide and multiplying by 100.
  • Percent Identity (Matches ⁇ 100)/Length of aligned region (with gaps).
  • the identity is calculated across the length of the contiguous nucleotide region. In embodiments where the entire contiguous nucleotide sequence of the oligonucleotide is the contiguous nucleotide region, identity is therefore calculated across the length of the nucleotide sequence of the oligonucleotide.
  • the contiguous nucleotide region may be identical to a region of the reference nucleic acid sequence, or in some embodiments may be identical to the entire reference nucleic acid. Unless otherwise indicated a sequence which has 100% identity to a reference sequence is referred to as being identical.
  • the reference sequence may be selected from the group consisting of any one of SEQ ID NOs 5-146 and 156.
  • the oligonucleotide comprises additional nucleotide(s) flanking the contiguous nucleotide region, for example region D′ or D′′, these additional flanking nucleotides may be disregarded when determining identity.
  • identity may be calculated across the entire oligonucleotide sequence.
  • the antisense oligonucleotide of the invention comprises a contiguous nucleotide region of 10-22 contiguous nucleotides which are identical to SEQ ID NO 156:
  • the contiguous nucleotide region consists or comprises of at least 10 contiguous nucleotides, such as 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, contiguous nucleotides, such as from 12-22, such as from 14-18 contiguous nucleotides of SEQ ID NO 156.
  • the entire contiguous sequence of the oligonucleotide consists or comprises of at least 10 contiguous nucleotides, such as 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, contiguous nucleotides, such as from 12-22, such as from 14-18 contiguous nucleotides of SEQ ID NO 156.
  • the contiguous nucleotide region is at least 12 contiguous nucleotides of SEQ ID NO 156. In some embodiments, the contiguous nucleotide region is at least 14 contiguous nucleotides of SEQ ID NO 156. In some embodiments, the contiguous nucleotide region is at least 16 contiguous nucleotides SEQ ID NO 156.
  • the contiguous nucleotide region is at least 10, 12, 14 or 16 contiguous nucleotides which are identical to SEQ ID NO 143.
  • the contiguous nucleotide region is at least 10, 12, 14 or 16 contiguous nucleotides which are identical to SEQ ID NO 145.
  • the contiguous nucleotide region is at least 10, 11, 12, 13, 14, 15 or 16 contiguous nucleotides which are identical to SEQ ID NO 143.
  • the contiguous nucleotide region is at least 10, 11, 12, 13, 14, 15, 16 or 17 contiguous nucleotides which are identical to SEQ ID NO 145.
  • the contiguous nucleotide consists or comprises SEQ ID NO 143.
  • the contiguous nucleotide region consists or comprises SEQ ID NO 145.
  • the contiguous nucleotide region is at least 10, 12, 14 or 16 contiguous nucleotides which are identical to a sequence selected from the group consisting of SEQ ID NO 138, 139, 140, 141, 142, 143, 144 and 145. In some embodiments, the contiguous nucleotide region comprises or consists of a sequence selected from the group consisting of SEQ ID NO 138, 139, 140, 141, 142, 143, 144 and 145.
  • the contiguous nucleotide region comprises the sequence SEQ ID NO 146: TTTACCTGGTT.
  • the invention provides for an antisense oligonucleotide 11-30 nucleotides in length, such as 12-20 nucleotides in length, which comprises the sequence SEQ ID NO 146: TTTACCTGGTT.
  • hybridizing or “hybridizes” as used herein is to be understood as two nucleic acid strands (e.g. an oligonucleotide and a target nucleic acid) forming hydrogen bonds between base pairs on opposite strands thereby forming a duplex.
  • the affinity of the binding between two nucleic acid strands is the strength of the hybridization. It is often described in terms of the melting temperature (T m ) defined as the temperature at which half of the oligonucleotides are duplexed with the target nucleic acid. At physiological conditions T m is not strictly proportional to the affinity (Mergny and Lacroix, 2003 , Oligonucleotides 13:515-537).
  • ⁇ G° is the energy associated with a reaction where aqueous concentrations are 1M, the pH is 7, and the temperature is 37° C.
  • ⁇ G° can be measured experimentally, for example, by use of the isothermal titration calorimetry (ITC) method as described in Hansen et al., 1965 , Chem. Comm. 36-38 and Holdgate et al., 2005, Drug Discov Today. The skilled person will know that commercial equipment is available for ⁇ G° measurements. ⁇ G° can also be estimated numerically by using the nearest neighbor model as described by SantaLucia, 1998 , Proc Natl Mad Sci USA.
  • ITC isothermal titration calorimetry
  • oligonucleotides of the present invention hybridize to a target nucleic acid with estimated ⁇ G° values below ⁇ 10 kcal for oligonucleotides that are 10-30 nucleotides in length.
  • the degree or strength of hybridization is measured by the standard state Gibbs free energy ⁇ G°.
  • the oligonucleotides may hybridize to a target nucleic acid with estimated ⁇ G° values below the range of ⁇ 10 kcal, such as below ⁇ 15 kcal, such as below ⁇ 20 kcal and such as below ⁇ 25 kcal for oligonucleotides that are 8-30 nucleotides in length.
  • the oligonucleotides hybridize to a target nucleic acid with an estimated ⁇ G° value of ⁇ 10 to ⁇ 60 kcal, such as ⁇ 12 to ⁇ 40, such as from ⁇ 15 to ⁇ 30 kcal or ⁇ 16 to ⁇ 27 kcal such as ⁇ 18 to ⁇ 25 kcal.
  • the oligonucleotide comprises a contiguous nucleotide region which is complementary to or hybridizes to a sub-sequence of the target nucleic acid molecule.
  • target sequence refers to a sequence of nucleotides present in the target nucleic acid which comprises the nucleobase sequence which is complementary to the contiguous nucleotide region or sequence of the oligonucleotide of the invention.
  • the target sequence consists of a region on the target nucleic acid which is complementary to the contiguous nucleotide region or sequence of the oligonucleotide of the invention.
  • the target sequence is longer than the complementary sequence of a single oligonucleotide, and may, for example represent a preferred region of the target nucleic acid which may be targeted by several oligonucleotides of the invention.
  • the oligonucleotide of the invention comprises a contiguous nucleotide region which is complementary to the target nucleic acid, such as a target sequence.
  • the oligonucleotide comprises a contiguous nucleotide region of at least 10 nucleotides which is complementary to or hybridizes to a target sequence present in the target nucleic acid molecule.
  • the contiguous nucleotide region (and therefore the target sequence) comprises of at least 10 contiguous nucleotides, such as 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, contiguous nucleotides, such as from 12-22, such as from 14-18 contiguous nucleotides.
  • the target sequence is, or is present within SEQ ID NO 147.
  • the target sequence is selected from the group consisting of SEQ ID NO 148, 149, 150, 151, 152, 153, 154 and 155:
  • SEQ ID NO 148 AACAACCAGGTAAATA SEQ ID NO 149: CAACCAGGTAAATATTTG SEQ ID NO 150: CCAACAACCAGGTAAA SEQ ID NO 151: AACCAGGTAAATATTTGG SEQ ID NO 152: ACAACCAGGTAAATATTTGG SEQ ID NO 153: CAACAACCAGGTAAATAT SEQ ID NO 154: ACAACCAGGTAAATAT SEQ ID NO 155: AACAACCAGGTAAATAT
  • the invention provides for an antisense oligonucleotide of 10-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of at least 10, contiguous nucleotides which are complementary to a sequence present in a sequence selected from SEQ ID NO 147 & 148-155.
  • the invention provides for an antisense oligonucleotide of 12-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of at least 12 contiguous nucleotides which are complementary to a sequence present in a sequence selected from SEQ ID NO 147 & 148-155.
  • the invention provides for an antisense oligonucleotide of 14-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of at least 14 contiguous nucleotides which are complementary to a sequence present in a sequence selected from SEQ ID NO 147 & 148-155.
  • the invention provides for an antisense oligonucleotide which consists or comprises a contiguous nucleotide region which is complementary to a sequence selected from SEQ ID NO 148-155.
  • the target sequence may be a sub-sequence of the target nucleic acid.
  • the oligonucleotide or contiguous nucleotide region is fully complementary to, or only comprises one or two mismatches to an HTRA1 sub-sequence, such as a sequence selected from the group consisting of SEQ ID NO 148-154.
  • the oligonucleotide or contiguous nucleotide region is fully complementary to, or only comprises one or two mismatches to an HTRA1 sub-sequence SEQ ID NO 147.
  • a target cell refers to a cell which is expressing the target nucleic acid.
  • the target cell may be in vivo or in vitro.
  • the target cell is a mammalian cell such as a rodent cell, such as a mouse cell, or a rat cell, or a primate cell such as a monkey cell or a human cell.
  • the cell may be a pig cell, a dog cell or a rabbit cell.
  • the target cell may be a retinal cell, such as a retinal pigment epithelium (PRE) cell.
  • PRE retinal pigment epithelium
  • the cell is selected from the group consisting of RPE cells, Bipolar Cell, Amacrine cells, Endothelial cells, Ganglion cells and Microglia cells.
  • the target cell may be a primary cell or an established cell line, such as U251, ARPE19, HEK293, or rat C6 cells.
  • the target nucleic acid is a nucleic acid which encodes mammalian HTRA1 and may for example be a gene, a RNA, a mRNA, and pre-mRNA, a mature mRNA or a cDNA sequence.
  • the target may therefore be referred to as an HTRA1 target nucleic acid.
  • the target nucleic acid encodes an HTRA1 protein, in particular mammalian HTRA1, such as human HTRA1 (See for example tables 1 & 2 which provides the mRNA and pre-mRNA sequences for human and rat HTRA1).
  • HTRA1 protein in particular mammalian HTRA1, such as human HTRA1 (See for example tables 1 & 2 which provides the mRNA and pre-mRNA sequences for human and rat HTRA1).
  • the target nucleic acid is selected from the group consisting of SEQ ID NO: 1, 2, 3, and 4, or naturally occurring variants thereof (e.g. sequences encoding a mammalian HTRA1 protein.
  • a target cell is a cell which is expressing the HTRA1 target nucleic acid.
  • the target nucleic acid is the HTRA1 mRNA, such as the HTRA1 pre-mRNA or HTRA1 mature mRNA.
  • the poly A tail of HTRA1 mRNA is typically disregarded for antisense oligonucleotide targeting.
  • the target nucleic acid may be a cDNA or a synthetic nucleic acid derived from DNA or RNA.
  • the target sequence may be a sub-sequence of the target nucleic acid.
  • the oligonucleotide or contiguous nucleotide region is fully complementary to, or only comprises one or two mismatches to an HTRA1 sub-sequence, such as a sequence selected from the group consisting of SEQ ID NO 148, 149, 150, 151, 152, 153, 154 and 155.
  • Complementarity to the target or sub-sequence thereof is measured over the length of the oligonucleotide, or contiguous nucleotide region thereof.
  • the oligonucleotide of the invention is typically capable of inhibiting the expression of the HTRA1 target nucleic acid in a cell which is expressing the HTRA1 target nucleic acid.
  • the contiguous sequence of nucleobases of the oligonucleotide of the invention is typically complementary to the HTRA1 target nucleic acid, as measured across the length of the oligonucleotide, optionally with the exception of one or two mismatches, and optionally excluding nucleotide based linker regions which may link the oligonucleotide to an optional functional group such as a conjugate, or other non-complementary terminal nucleotides (e.g. region D).
  • the target nucleic acid may, in some embodiments, be a RNA or DNA, such as a messenger RNA, such as a mature mRNA or a pre-mRNA.
  • the target nucleic acid is a RNA or DNA which encodes mammalian HTRA1 protein, such as human HTRA1, e.g. the human HTRA1 mRNA sequence, such as that disclosed as SEQ ID NO 1 (NM_002775.4, GI:190014575). Further information on exemplary target nucleic acids is provided in tables 1 & 2.
  • the genome coordinates provide the pre-mRNA sequence (genomic sequence).
  • the NCBI reference provides the mRNA sequence (cDNA sequence). *The National Center for Biotechnology Information reference sequence database is a comprehensive, integrated, non-redundant, well-annotated set of reference sequences including genomic, transcript, and protein. It is hosted at www.ncbi.nlm.nih.gov/refseq.
  • naturally occurring variant refers to variants of HTRA1 gene or transcripts which originate from the same genetic loci as the target nucleic acid, but may differ for example, by virtue of degeneracy of the genetic code causing a multiplicity of codons encoding the same amino acid, or due to alternative splicing of pre-mRNA, or the presence of polymorphisms, such as single nucleotide polymorphisms, and allelic variants. Based on the presence of the sufficient complementary sequence to the oligonucleotide, the oligonucleotide of the invention may therefore target the target nucleic acid and naturally occurring variants thereof.
  • the naturally occurring variants have at least 95% such as at least 98% or at least 99% homology to a mammalian HTRA1 target nucleic acid, such as a target nucleic acid selected form the group consisting of SEQ ID NO 1, 2, 3, or 4.
  • modulation of expression is to be understood as an overall term for an oligonucleotide's ability to alter the amount of HTRA1 when compared to the amount of HTRA1 before administration of the oligonucleotide. Alternatively modulation of expression may be determined by reference to a control experiment where the oligonucleotide of the invention is not administered.
  • One type of modulation is an oligonucleotide's ability to inhibit, down-regulate, reduce, suppress, remove, stop, block, prevent, lessen, lower, avoid or terminate expression of HTRA1, e.g. by degradation of mRNA or blockage of transcription.
  • the antisense oligonucleotide of the invention are capable of inhibiting, down-regulating, reduce, suppress, remove, stop, block, prevent, lessen, lower, avoid or terminate expression of HTRA1.
  • a high affinity modified nucleoside is a modified nucleotide which, when incorporated into the oligonucleotide enhances the affinity of the oligonucleotide for its complementary target, for example as measured by the melting temperature (T m ).
  • a high affinity modified nucleoside of the present invention preferably result in an increase in melting temperature between +0.5 to +12° C., more preferably between +1.5 to +10° C. and most preferably between +3 to +8° C. per modified nucleoside.
  • Numerous high affinity modified nucleosides are known in the art and include for example, many 2′ substituted nucleosides as well as locked nucleic acids (LNA) (see e.g. Freier & Altmann; Nucl. Acid Res., 1997, 25, 4429-4443 and Uhlmann; Curr. Opinion in Drug Development, 2000, 3(2), 293-213).
  • the oligomer of the invention may comprise one or more nucleosides which have a modified sugar moiety, i.e. a modification of the sugar moiety when compared to the ribose sugar moiety found in DNA and RNA.
  • nucleosides with modification of the ribose sugar moiety have been made, primarily with the aim of improving certain properties of oligonucleotides, such as affinity and/or nuclease resistance.
  • Such modifications include those where the ribose ring structure is modified, e.g. by replacement with a hexose ring (HNA), or a bicyclic ring, which typically have a biradicle bridge between the C2 and C4 carbons on the ribose ring (LNA), or an unlinked ribose ring which typically lacks a bond between the C2 and C3 carbons (e.g. UNA).
  • HNA hexose ring
  • LNA ribose ring
  • UPA unlinked ribose ring which typically lacks a bond between the C2 and C3 carbons
  • Other sugar modified nucleosides include, for example, bicyclohexose nucleic acids (WO2011/017521) or tricyclic nucleic acids (WO2013/154798). Modified nucleosides also include nucleosides where the sugar moiety is replaced with a non-sugar moiety, for example in the case of
  • Sugar modifications also include modifications made via altering the substituent groups on the ribose ring to groups other than hydrogen, or the 2′—OH group naturally found in DNA and RNA nucleosides. Substituents may, for example be introduced at the 2′, 3′, 4′ or 5′ positions. Nucleosides with modified sugar moieties also include 2′ modified nucleosides, such as 2′ substituted nucleosides. Indeed, much focus has been spent on developing 2′ substituted nucleosides, and numerous 2′ substituted nucleosides have been found to have beneficial properties when incorporated into oligonucleotides, such as enhanced nucleoside resistance and enhanced affinity.
  • a 2′ sugar modified nucleoside is a nucleoside which has a substituent other than H or —OH at the 2′ position (2′ substituted nucleoside) or comprises a 2′ linked biradicle, and includes 2′ substituted nucleosides and LNA (2′-4′ biradicle bridged) nucleosides.
  • the 2′ modified sugar may provide enhanced binding affinity and/or increased nuclease resistance to the oligonucleotide.
  • 2′ substituted modified nucleosides are 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA (MOE), 2′-amino-DNA, 2′-Fluoro-RNA, and 2′-F-ANA nucleoside.
  • MOE 2′-O-methoxyethyl-RNA
  • 2′-amino-DNA 2′-Fluoro-RNA
  • 2′-F-ANA nucleoside examples of 2′ substituted modified nucleosides.
  • LNA Locked Nucleic Acid Nucleosides
  • LNA nucleosides are modified nucleosides which comprise a linker group (referred to as a biradicle or a bridge) between C2′ and C4′ of the ribose sugar ring of a nucleotide. These nucleosides are also termed bridged nucleic acid or bicyclic nucleic acid (BNA) in the literature.
  • a linker group referred to as a biradicle or a bridge
  • the modified nucleoside or the LNA nucleosides of the oligomer of the invention has a general structure of the formula I or II:
  • W is selected from —O—, —S—, —N(R a )—, —C(R a R b )—, such as, in some embodiments —O—;
  • Z designates an internucleoside linkage to an adjacent nucleoside, or a 5′-terminal group
  • Z* designates an internucleoside linkage to an adjacent nucleoside, or a 3′-terminal group
  • X designates a group selected from the list consisting of —C(R a R b )—, —C(R a ) ⁇ C(R b )—, —C(R a ) ⁇ N—, —O—, —Si(R a ) 2 —, —S—, —SO 2 —, —N(R a )—, and >C ⁇ Z
  • X is selected from the group consisting of: —O—, —S—, NH—, NR a R b , —CH 2 —, CR a R b , —C( ⁇ CH 2 )—, and —C( ⁇ CR a R b )—
  • Y designates a group selected from the group consisting of —C(R a R b )—, —C(R a ) ⁇ C(R b )—, —C(R a ) ⁇ N—, —O—, —Si(R a ) 2 —, —S—, —SO 2 —, —N(R a )—, and >C ⁇ Z
  • Y is selected from the group consisting of: —CH 2 —, —C(R a R b )—, —CH 2 CH 2 —, —C(R a R b )—C(R a R b )—, —CH 2 CH 2 CH 2 —, —C(R a R b )C(R a R b )C(R a R b )—, —C(R a ) ⁇ C(R b )—, and —C(R a ) ⁇ N—
  • Y is selected from the group consisting of: —CH 2 —, —CHR a —, —CHCH 3 —, CR a R b —
  • bivalent linker group also referred to as a radicle
  • a bivalent linker group consisting of 1, 2, or 3 groups/atoms selected from the group consisting of —C(R a R b )—, —C(R a ) ⁇ C(R b )—, —C(R a ) ⁇ N—, —O—, —Si(R a ) 2 —, —S—, —SO 2 —, —N(R a )—, and >C ⁇ Z
  • —X—Y— designates a biradicle selected from the groups consisting of: —X—CH 2 —, —X—CR a R b —, —X—CHR a ⁇ , —X—C(HCH 3 ) ⁇ , —O—Y—, —O—CH 2 —, —S—CH 2 —, —NH—CH 2 —, —O—CHCH 3 —, —CH 2 —O—CH 2 , —O—CH(CH 3 CH 3 )—, —O—CH 2 —CH 2 —, OCH 2 —CH 2 —CH 2 —, —O—CH 2 OCH 2 —, —O—NCH 2 —, —C( ⁇ CH 2 )—CH 2 —, —NR a —CH 2 —, N—O—CH 2 , —S—CR a R b — and —S—CHR a —.
  • Z is selected from —O—, —S—, and —N(R a )—
  • R a and R a and, when present R b each is independently selected from hydrogen, optionally substituted C 1-6 -alkyl, optionally substituted C 2-6 -alkenyl, optionally substituted C 2-6 -alkynyl, hydroxy, optionally substituted C 1-6 -alkoxy, C 2-6 -alkoxyalkyl, C 2-6 -alkenyloxy, carboxy, C 1-6 -alkoxycarbonyl, C 1-6 -alkylcarbonyl, formyl, aryl, aryloxy-carbonyl, aryloxy, arylcarbonyl, heteroaryl, heteroaryloxy-carbonyl, heteroaryloxy, heteroarylcarbonyl, amino, mono- and di(C 1-6 -alkyl)amino, carbamoyl, mono- and di(C 1-6 -alkyl)-amino-carbonyl, amino-C 1-6 -alkyl-aminocarbonyl, mono- and di
  • R 1 , R 2 , R 3 , R 5 and R 5* are independently selected from the group consisting of: hydrogen, optionally substituted C 1-6 -alkyl, optionally substituted C 2-6 -alkenyl, optionally substituted C 2-6 -alkynyl, hydroxy, C 1-6 -alkoxy, C 2-6 -alkoxyalkyl, C 2-6 -alkenyloxy, carboxy, C 1-6 -alkoxycarbonyl, C 1-6 -alkylcarbonyl, formyl, aryl, aryloxy-carbonyl, aryloxy, arylcarbonyl, heteroaryl, heteroaryloxy-carbonyl, heteroaryloxy, heteroarylcarbonyl, amino, mono- and di(C 1-6 -alkyl)amino, carbamoyl, mono- and di(C 1-6 -alkyl)-amino-carbonyl, amino-C 1-6 -alkyl-aminocarbonyl
  • R 1 , R 2 , R 3 , R 5 and R 5* are independently selected from C 1-6 alkyl, such as methyl, and hydrogen.
  • R 1 , R 2 , R 3 are all hydrogen, and either R 5 and R 5* is also hydrogen and the other of R 5 and R 5* is other than hydrogen, such as C 1-6 alkyl such as methyl.
  • R a is either hydrogen or methyl. In some embodiments, when present, R b is either hydrogen or methyl.
  • one of R a and R b is hydrogen and the other is other than hydrogen
  • the biradicle —X—Y— is —O—CH 2 —
  • W is O
  • all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • LNA nucleosides are disclosed in WO99/014226, WO00/66604, WO98/039352 and WO2004/046160 which are all hereby incorporated by reference, and include what are commonly known as beta-D-oxy LNA and alpha-L-oxy LNA nucleosides.
  • the biradicle —X—Y— is —S—CH 2 —
  • W is O
  • all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • Such thio LNA nucleosides are disclosed in WO99/014226 and WO2004/046160 which are hereby incorporated by reference.
  • the biradicle —X—Y— is —NH—CH 2 —
  • W is O
  • all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • amino LNA nucleosides are disclosed in WO99/014226 and WO2004/046160 which are hereby incorporated by reference.
  • the biradicle —X—Y— is —O—CH 2 —CH 2 — or —O—CH 2 —CH 2 —CH 2 —
  • W is O
  • all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • LNA nucleosides are disclosed in WO00/047599 and Morita et al, Bioorganic & Med. Chem. Lett. 12 73-76, which are hereby incorporated by reference, and include what are commonly known as 2′-O-4′C-ethylene bridged nucleic acids (ENA).
  • the biradicle —X—Y— is —O—CH 2 —
  • W is O
  • all of R 1 , R 2 , R 3 , and one of R 5 and R 5* are hydrogen
  • the other of R 5 and R 5* is other than hydrogen such as C 1-6 alkyl, such as methyl.
  • the biradicle —X—Y— is —O—CR a R b —, wherein one or both of R a and R b are other than hydrogen, such as methyl, W is O, and all of R 1 , R 2 , R 3 , and one of R 5 and R 5* are hydrogen, and the other of R 5 and R 5* is other than hydrogen such as C 1-6 alkyl, such as methyl.
  • R a and R b are other than hydrogen, such as methyl
  • W is O
  • all of R 1 , R 2 , R 3 , and one of R 5 and R 5* are hydrogen
  • the other of R 5 and R 5* is other than hydrogen such as C 1-6 alkyl, such as methyl.
  • the biradicle —X—Y— designate the bivalent linker group —O—CH(CH 2 OCH 3 )— (2′ O-methoxyethyl bicyclic nucleic acid—Seth at al., 2010, J. Org. Chem. Vol 75(5) pp. 1569-81).
  • the biradicle —X—Y— designate the bivalent linker group —O—CH(CH 2 CH 3 )— (2′O-ethyl bicyclic nucleic acid—Seth at al., 2010, J. Org. Chem. Vol 75(5) pp. 1569-81).
  • the biradicle —X—Y— is —O—CHR a —
  • W is O
  • all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • 6′ substituted LNA nucleosides are disclosed in WO10036698 and WO07090071 which are both hereby incorporated by reference.
  • the biradicle —X—Y— is —O—CH(CH 2 OCH 3 )—, W is O, and all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • LNA nucleosides are also known as cyclic MOEs in the art (cMOE) and are disclosed in WO07090071.
  • the biradicle —X—Y— designate the bivalent linker group —O—CH(CH 3 )—. — in either the R— or S— configuration. In some embodiments, the biradicle —X—Y— together designate the bivalent linker group —O—CH 2 —O—CH 2 — (Seth at al., 2010, J. Org. Chem). In some embodiments, the biradicle —X—Y— is —O—CH(CH 3 )—, W is O, and all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • Such 6′ methyl LNA nucleosides are also known as cET nucleosides in the art, and may be either (S)cET or (R)cET stereoisomers, as disclosed in WO07090071 (beta-D) and WO2010/036698 (alpha-L) which are both hereby incorporated by reference).
  • the biradicle —X—Y— is —O—CR a R b —, wherein in neither R a or R b is hydrogen, W is O, and all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • R a and R b are both methyl.
  • the biradicle —X—Y— is —S—CHR a —
  • W is O
  • all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • R a is methyl.
  • the biradicle —X—Y— is —C( ⁇ CH 2 )—C(R a R b )—, such as —C( ⁇ CH 2 )—CH 2 —, or —C( ⁇ CH 2 )—CH(CH 3 )—W is O, and all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • vinyl carbo LNA nucleosides are disclosed in WO08154401 and WO09067647 which are both hereby incorporated by reference.
  • the biradicle —X—Y— is —N(—OR a )—, W is O, and all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • R a is C 1-6 alkyl such as methyl.
  • LNA nucleosides are also known as N substituted LNAs and are disclosed in WO2008/150729 which is hereby incorporated by reference.
  • the biradicle —X—Y— together designate the bivalent linker group —O—NR a —CH 3 — (Seth at al., 2010, J. Org. Chem).
  • the biradicle —X—Y— is —N(R a )—, W is O, and all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • R a is C 1-6 alkyl such as methyl.
  • R 5 and R 5* is hydrogen and, when substituted the other of R 5 and R 5* is C 1-6 alkyl such as methyl.
  • R 1 , R 2 , R 3 may all be hydrogen, and the biradicle —X—Y— may be selected from —O—CH 2 — or —O—C(HCR a )—, such as —O—C(HCH 3 )—.
  • the biradicle is —CR a R b —O—CR a R b —, such as CH 2 —O—CH 2 —, W is O and all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • R a is C 1-6 alkyl such as methyl.
  • LNA nucleosides are also known as conformationally restricted nucleotides (CRNs) and are disclosed in WO2013036868 which is hereby incorporated by reference.
  • the biradicle is —O—CR a R b —O—CR a R b —, such as O—CH 2 —O—CH 2 —, W is O and all of R 1 , R 2 , R 3 , R 5 and R 5* are all hydrogen.
  • R a is C 1-6 alkyl such as methyl.
  • LNA nucleosides are also known as COC nucleotides and are disclosed in Mitsuoka et al., Nucleic Acids Research 2009 37(4), 1225-1238, which is hereby incorporated by reference.
  • the LNA nucleosides may be in the beta-D or alpha-L stereoisoform.
  • the LNA nucleosides in the oligonucleotides are beta-D-oxy-LNA nucleosides.
  • Nuclease mediated degradation refers to an oligonucleotide capable of mediating degradation of a complementary nucleotide sequence when forming a duplex with such a sequence.
  • the oligonucleotide may function via nuclease mediated degradation of the target nucleic acid, where the oligonucleotides of the invention are capable of recruiting a nuclease, particularly and endonuclease, preferably endoribonuclease (RNase), such as RNase H.
  • RNase endoribonuclease
  • oligonucleotide designs which operate via nuclease mediated mechanisms are oligonucleotides which typically comprise a region of at least 5 or 6 DNA nucleosides and are flanked on one side or both sides by affinity enhancing nucleosides, for example gapmers, headmers and tailmers.
  • the RNase H activity of an antisense oligonucleotide refers to its ability to recruit RNase H when in a duplex with a complementary RNA molecule.
  • WO01/23613 provides in vitro methods for determining RNaseH activity, which may be used to determine the ability to recruit RNaseH.
  • an oligonucleotide is deemed capable of recruiting RNase H if it, when provided with a complementary target nucleic acid sequence, has an initial rate, as measured in pmol/l/min, of at least 5%, such as at least 10% or more than 20% of the of the initial rate determined when using a oligonucleotide having the same base sequence as the modified oligonucleotide being tested, but containing only DNA monomers, with phosphorothioate linkages between all monomers in the oligonucleotide, and using the methodology provided by Example 91-95 of WO01/23613 (hereby incorporated by reference).
  • gapmer refers to an antisense oligonucleotide which comprises a region of RNase H recruiting oligonucleotides (gap) which is flanked 5′ and 3′ by regions which comprise one or more affinity enhancing modified nucleosides (flanks or wings).
  • oligonucleotides capable of recruiting RNase H where one of the flanks is missing, i.e. only one of the ends of the oligonucleotide comprises affinity enhancing modified nucleosides.
  • the 3′ flank is missing (i.e. the 5′ flank comprises affinity enhancing modified nucleosides) and for tailmers the 5′ flank is missing (i.e. the 3′ flank comprises affinity enhancing modified nucleosides).
  • LNA gapmer is a gapmer oligonucleotide wherein at least one of the affinity enhancing modified nucleosides is an LNA nucleoside.
  • mixed wing gapmer refers to a LNA gapmer wherein the flank regions comprise at least one LNA nucleoside and at least one non-LNA modified nucleoside, such as at least one DNA nucleoside or at least one 2′ substituted modified nucleoside, such as, for example, 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA (MOE), 2′-amino-DNA, 2′-Fluoro-RNA and 2′-F-ANA nucleoside(s).
  • the mixed wing gapmer has one flank which comprises LNA nucleosides (e.g. 5′ or 3′) and the other flank (3′ or 5′ respectfully) comprises 2′ substituted modified nucleoside(s).
  • conjugate refers to an oligonucleotide which is covalently linked to a non-nucleotide moiety (conjugate moiety or region C or third region).
  • conjugate refers to an oligonucleotide which is covalently linked to a non-nucleotide moiety (conjugate moiety or region C or third region).
  • the non-nucleotide moiety selected from the group consisting of a protein, such as an enzyme, an antibody or an antibody fragment or a peptide; a lipophilic moiety such as a lipid, a phospholipid, a sterol; a polymer, such as polyethyleneglycol or polypropylene glycol; a receptor ligand; a small molecule; a reporter molecule; and a non-nucleosidic carbohydrate.
  • a linkage or linker is a connection between two atoms that links one chemical group or segment of interest to another chemical group or segment of interest via one or more covalent bonds.
  • Conjugate moieties can be attached to the oligonucleotide directly or through a linking moiety (e.g. linker or tether).
  • Linkers serve to covalently connect a third region, e.g. a conjugate moiety to an oligonucleotide (e.g. the termini of region A or C).
  • the conjugate or oligonucleotide conjugate of the invention may optionally, comprise a linker region which is positioned between the oligonucleotide and the conjugate moiety.
  • the linker between the conjugate and oligonucleotide is biocleavable.
  • Biocleavable linkers comprising or consisting of a physiologically labile bond that is cleavable under conditions normally encountered or analogous to those encountered within a mammalian body.
  • Conditions under which physiologically labile linkers undergo chemical transformation include chemical conditions such as pH, temperature, oxidative or reductive conditions or agents, and salt concentration found in or analogous to those encountered in mammalian cells.
  • Mammalian intracellular conditions also include the presence of enzymatic activity normally present in a mammalian cell such as from proteolytic enzymes or hydrolytic enzymes or nucleases.
  • the biocleavable linker is susceptible to S1 nuclease cleavage.
  • the nuclease susceptible linker comprises between 1 and 10 nucleosides, such as 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleosides, more preferably between 2 and 6 nucleosides and most preferably between 2 and 4 linked nucleosides comprising at least two consecutive phosphodiester linkages, such as at least 3 or 4 or 5 consecutive phosphodiester linkages.
  • the nucleosides are DNA or RNA.
  • Phosphodiester containing biocleavable linkers are described in more detail in WO 2014/076195 (hereby incorporated by reference), and may be referred to as region D herein.
  • Conjugates may also be linked to the oligonucleotide via non biocleavable linkers, or in some embodiments the conjugate may comprise a non-cleavable linker which is covalently attached to the biocleavable linker.
  • Linkers that are not necessarily biocleavable but primarily serve to covalently connect a conjugate moiety to an oligonucleotide or biocleavable linker.
  • Such linkers may comprise a chain structure or an oligomer of repeating units such as ethylene glycol, amino acid units or amino alkyl groups.
  • the linker (region Y) is an amino alkyl, such as a C 2 -C 36 amino alkyl group, including, for example C 6 to C 12 amino alkyl groups. In some embodiments the linker (region Y) is a C 6 amino alkyl group. Conjugate linker groups may be routinely attached to an oligonucleotide via use of an amino modified oligonucleotide, and an activated ester group on the conjugate group.
  • treatment refers to both treatment of an existing disease (e.g. a disease or disorder as herein referred to), or prevention of a disease, i.e. prophylaxis. It will therefore be recognized that treatment as referred to herein may, in some embodiments, be prophylactic.
  • the invention relates to oligonucleotides capable of inhibiting the expression of HTRA1.
  • the modulation is may achieved by hybridizing to a target nucleic acid encoding HTRA1 or which is involved in the regulation of HTRA1.
  • the target nucleic acid may be a mammalian HTRA 1 sequence, such as a sequence selected from the group consisting of SEQ ID 1, 2, 3 or 4.
  • the oligonucleotide of the invention is an antisense oligonucleotide which targets HTRA1, such as a mammalian HTRA1.
  • the antisense oligonucleotide of the invention is capable of modulating the expression of the target by inhibiting or down-regulating it. Preferably, such modulation produces an inhibition of expression of at least 20% compared to the normal expression level of the target, such as at least 30%, 40%, 50%, 60%, 70%, 80%, or 90% inhibition compared to the normal expression level of the target.
  • compounds of the invention may be capable of inhibiting expression levels of HTRA1 mRNA by at least 60% or 70% in vitro using ARPE-19 cells. In some embodiments compounds of the invention may be capable of inhibiting expression levels of HTRA1 mRNA by at least 60% or 70% in vitro using ARPE-19 cells.
  • compounds of the invention may be capable of inhibiting expression levels of HTRA1 protein by at least 50% in vitro using ARPE-19 cells.
  • the examples provide assays which may be used to measure HTRA1 RNA or protein inhibition (e.g. example 3).
  • the target modulation is triggered by the hybridization between a contiguous nucleotide sequence of the oligonucleotide and the target nucleic acid.
  • the oligonucleotide of the invention comprises mismatches between the oligonucleotide and the target nucleic acid. Despite mismatches hybridization to the target nucleic acid may still be sufficient to show a desired modulation of HTRA1 expression.
  • Reduced binding affinity resulting from mismatches may advantageously be compensated by increased number of nucleotides in the oligonucleotide and/or an increased number of modified nucleosides capable of increasing the binding affinity to the target, such as 2′ modified nucleosides, including LNA, present within the oligonucleotide sequence.
  • An aspect of the present invention relates to an antisense oligonucleotide which comprises a contiguous nucleotide region of 10 to 30 nucleotides in length with at least 90% complementarity to HTRA1 target sequence, such as fully complementary to an HTRA1 target sequence, e.g. a nucleic acid selected from the group consisting SEQ ID NO 1, 2, 3 & 4.
  • the oligonucleotide comprises a contiguous sequence which is at least 90% complementary, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, or 100% complementary with a region of the target nucleic acid.
  • the oligonucleotide of the invention or a contiguous nucleotide sequence thereof is fully complementary (100% complementary) to a region of the target nucleic acid, or in some embodiments may comprise one or two mismatches between the oligonucleotide and the target nucleic acid.
  • the oligonucleotide, or a contiguous nucleotide sequence of at least 12 nucleotides thereof is at least 90% complementary, such as fully (or 100%) complementary to a region of SEQ ID NO 147.
  • the oligonucleotide, or a contiguous nucleotide sequence of at least 12 nucleotides thereof is at least 90% complementary, such as fully (or 100%) complementary to a region of a sequence selected from the group consisting of SEQ ID NOs 148, 149, 150, 151, 152, 153, 154 and 155.
  • the oligonucleotide, or a contiguous nucleotide sequence of at least 14 nucleotides thereof is fully (or 100%) complementary to SEQ ID 147, or a sequence selected from the group consisting of SEQ ID NOs 148, 149, 150, 151, 152, 153, 154 and 155.
  • the oligonucleotide, or a contiguous nucleotide sequence of at least 16 nucleotides thereof is fully (or 100%) complementary to SEQ ID 147, or a sequence selected from the group consisting of SEQ ID NOs 148, 149, 150, 151, 152, 153, 154 and 155.
  • the oligonucleotide, or contiguous nucleotide region thereof is fully (or 100%) complementary to a sequence selected from the group consisting of SEQ ID NOs 148, 149, 150, 151, 152, 153, 154 and 155.
  • the oligonucleotide or contiguous nucleotide region thereof comprises or consists of a sequence selected from the group consisting of SEQ ID NOs 143, 138, 139, 140, 141, 142, 144 and 145:
  • SEQ ID NO 143 TATTTACCTGGTTGTT SEQ ID NO 138: CAAATATTTACCTGGTTG SEQ ID NO 139: TTTACCTGGTTGTTGG SEQ ID NO 140: CCAAATATTTACCTGGTT SEQ ID NO 141: CCAAATATTTACCTGGTTGT SEQ ID NO 142: ATATTTACCTGGTTGTTG SEQ ID NO 144: ATATTTACCTGGTTGT SEQ ID NO 145: ATATTTACCTGGTTGTT
  • oligonucleotide motif sequences can be modified to for example increase nuclease resistance and/or binding affinity to the target nucleic acid. Modifications are described in the definitions and in the “Oligonucleotide design” section.
  • the oligonucleotide of the invention, or contiguous nucleotide region thereof is fully complementary (100% complementary) to a region of the target nucleic acid, or in some embodiments may comprise one or two mismatches between the oligonucleotide and the target nucleic acid.
  • the oligonucleotide, or contiguous nucleotide sequence of at least 12 nucleotides thereof is at least 90% complementary, such as fully (or 100%) complementary to the target nucleic acid sequence.
  • the oligonucleotide, or a contiguous nucleotide sequence of at least 12 nucleotides thereof has 100% identity to a sequence selected from the group consisting of SEQ ID NOs 5-107, or SEQ ID NOs 108-137.
  • the oligonucleotide, or a contiguous nucleotide sequence of at least 14 nucleotides thereof has 100% identity to a sequence selected from the group consisting of SEQ ID NOs 5-107, or SEQ ID NOs 108-137.
  • the oligonucleotide, or contiguous nucleotide sequence of at least 16 nucleotides thereof has 100% identity to a sequence selected from the group consisting of SEQ
  • the oligonucleotide, or contiguous nucleotide region thereof comprises or consists of a sequence selected from SEQ ID NOs 5-107, or SEQ ID NOs 108-137.
  • Oligonucleotide design refers to the pattern of nucleoside sugar modifications in the oligonucleotide sequence.
  • the oligonucleotides of the invention comprise sugar-modified nucleosides and may also comprise DNA or RNA nucleosides.
  • the oligonucleotide comprises sugar-modified nucleosides and DNA nucleosides. Incorporation of modified nucleosides into the oligonucleotide of the invention may enhance the affinity of the oligonucleotide for the target nucleic acid. In that case, the modified nucleosides can be referred to as affinity enhancing modified nucleotides.
  • the oligonucleotide comprises at least 1 modified nucleoside, such as at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15 or at least 16 modified nucleosides.
  • the oligonucleotide comprises from 1 to 10 modified nucleosides, such as from 2 to 9 modified nucleosides, such as from 3 to 8 modified nucleosides, such as from 4 to 7 modified nucleosides, such as 6 or 7 modified nucleosides.
  • the oligonucleotide of the invention may comprise modifications, which are independently selected from these three types of modifications (modified sugar, modified nucleobase and modified internucleoside linkage) or a combination thereof.
  • the oligonucleotide comprises one or more sugar modified nucleosides, such as 2′ sugar modified nucleosides.
  • the oligonucleotide of the invention comprise the one or more 2′ sugar modified nucleoside independently selected from the group consisting of 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA, 2′-amino-DNA, 2′-fluoro-DNA, arabino nucleic acid (ANA), 2′-fluoro-ANA and LNA nucleosides. Even more preferably the one or more modified nucleoside is LNA.
  • At least 1 of the modified nucleosides is a locked nucleic acid (LNA), such as at least 2, such as at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 of the modified nucleosides are LNA. In a still further embodiment all the modified nucleosides are LNA.
  • LNA locked nucleic acid
  • the oligonucleotide comprises at least one modified internucleoside linkage.
  • the internucleoside linkages within the contiguous nucleotide sequence are phosphorothioate or boranophosphate internucleoside linkages. In some embodiments all the internucleotide linkages in the contiguous sequence of the oligonucleotide are phosphorothioate linkages.
  • the oligonucleotide of the invention comprise at least one modified nucleoside which is a 2′-MOE-RNA, such as 2, 3, 4, 5, 6, 7, 8, 9 or 10 2′-MOE-RNA nucleoside units.
  • at least one of said modified nucleoside is 2′-fluoro DNA, such as 2, 3, 4, 5, 6, 7, 8, 9 or 10 2′-fluoro-DNA nucleoside units.
  • the oligonucleotide of the invention comprises at least one LNA unit, such as 1, 2, 3, 4, 5, 6, 7, or 8 LNA units, such as from 2 to 6 LNA units, such as from 3 to 7 LNA units, 4 to 8 LNA units or 3, 4, 5, 6 or 7 LNA units.
  • all the modified nucleosides are LNA nucleosides.
  • all LNA cytosine units are 5-methyl-cytosine.
  • the oligonucleotide or contiguous nucleotide region thereof has at least 1 LNA unit at the 5′ end and at least 2 LNA units at the 3′ end of the nucleotide sequence.
  • all cytosine nucleobases present in the oligonucleotide of the invention are 5-methyl-cytosine.
  • the oligonucleotide of the invention comprises at least one LNA unit and at least one 2′ substituted modified nucleoside.
  • the oligonucleotide comprise both 2′ sugar modified nucleosides and DNA units.
  • the oligonucleotide of the invention is capable of recruiting RNase H.
  • the oligonucleotide of the invention or contiguous nucleotide region thereof is a gapmers oligonucleotide.
  • the oligonucleotide of the invention has a gapmer design or structure also referred herein merely as “Gapmer”.
  • Gapmer the oligonucleotide comprises at least three distinct structural regions a 5′-flank, a gap and a 3′-flank, F-G-F′ in ‘5->3’ orientation.
  • flanking regions F and F′ comprise at least one sugar modified nucleoside which is adjacent to region G, and may in some embodiments comprise a contiguous stretch of 2-7 sugar modified nucleoside, or a contiguous stretch of sugar modified and DNA nucleosides (mixed wings comprising both sugar modified and DNA nucleosides). Consequently, the nucleosides of the 5′ flanking region and the 3′ flanking region which are adjacent to the gap region are sugar modified nucleosides, such as 2′ modified nucleosides.
  • the gap region, G comprises a contiguous stretch of nucleotides which are capable of recruiting RNase H, when the oligonucleotide is in duplex with the HTRA1target nucleic acid.
  • region G comprises a contiguous stretch of 5-16 DNA nucleosides.
  • the gapmer region F-G-F′ is complementary to the HTRA1 target nucleic acid, and may therefore be the contiguous nucleotide region of the oligonucleotide.
  • Regions F and F′, flanking the 5′ and 3′ ends of region G may comprise one or more affinity enhancing modified nucleosides.
  • the 3′ flank comprises at least one LNA nucleoside, preferably at least 2 LNA nucleosides.
  • the 5′ flank comprises at least one LNA nucleoside.
  • both the 5′ and 3′ flanking regions comprise a LNA nucleoside.
  • all the nucleosides in the flanking regions are LNA nucleosides.
  • flanking regions may comprise both LNA nucleosides and other nucleosides (mixed flanks), such as DNA nucleosides and/or non-LNA modified nucleosides, such as 2′ substituted nucleosides.
  • the gap is defined as a contiguous sequence of at least 5 RNase H recruiting nucleosides (such as 5-16 DNA nucleosides) flanked at the 5′ and 3′ end by an affinity enhancing modified nucleoside, such as an LNA, such as beta-D-oxy-LNA.
  • Region F (5′ flank or 5′ wing) attached to the ′5 end of region G comprises, contains or consists of at least one sugar modified nucleoside such as at least 2, at least 3, at least 4, at least 5, at least 6, at least 7 modified nucleosides.
  • region F comprises or consists of from 1 to 7 modified nucleosides, such as from 2 to 6 modified nucleosides, such as from 2 to 5 modified nucleosides, such as from 2 to 4 modified nucleosides, such as from 1 to 3 modified nucleosides, such as 1, 2, 3 or 4 modified nucleosides.
  • one or more or all of the modified nucleosides in region F are 2′ modified nucleosides.
  • one or more of the 2′ modified nucleosides in region F are selected from 2′-O-alkyl-RNA units, 2′-O-methyl-RNA, 2′-amino-DNA units, 2′-fluoro-DNA units, 2′-alkoxy-RNA, MOE units, LNA units, arabino nucleic acid (ANA) units and 2′-fluoro-ANA units.
  • all the modified nucleosides in region F are LNA nucleosides.
  • the LNA nucleosides in region F are independently selected from the group consisting of oxy-LNA, thio-LNA, amino-LNA, cET, and/or ENA, in either the beta-D or alpha-L configurations or combinations thereof.
  • region F has at least 1 beta-D-oxy LNA unit, at the 5′ end of the contiguous sequence.
  • Region G may comprise, contain or consist of at 5-16 consecutive DNA nucleosides capable of recruiting RNaseH.
  • region G comprise, contain or consist of from 5 to 12, or from 6 to 10 or from 7 to 9, such as 8 consecutive nucleotide units capable of recruiting RNaseH.
  • At least one nucleoside unit in region G is a DNA nucleoside unit, such as from 4 to 20 or 6 to 18 DNA units, such as 5 to 16, In some embodiments, all of the nucleosides of region G are DNA units.
  • region G may consist of a mixture of DNA and other nucleosides capable of mediating RNase H cleavage.
  • at least 50% of the nucleosides of region G are DNA, such as at least 60%, at least 70% or at least 80%, or at least 90% DNA.
  • Region F′ (3′ flank or 3′ wing) attached to the ′3 end of region G comprises, contains or consists of at least one sugar modified nucleoside such as at least 2, at least 3, at least 4, at least 5, at least 6, at least 7 modified nucleosides.
  • region F′ comprises or consists of from 1 to 7 modified nucleosides, such as from 2 to 6 modified nucleosides, such as from 2 to 5 modified nucleosides, such as from 2 to 4 modified nucleosides, such as from 1 to 3 modified nucleosides, such as 1, 2, 3 or 4 modified nucleosides.
  • one or more or all of the modified nucleosides in region F′ are 2′ modified nucleosides.
  • one or more of the 2′ modified nucleosides in region F′ are selected from 2′-O-alkyl-RNA units, 2′-O-methyl-RNA, 2′-amino-DNA units, 2′-fluoro-DNA units, 2′-alkoxy-RNA, MOE units, LNA units, arabino nucleic acid (ANA) units and 2′-fluoro-ANA units.
  • all the modified nucleosides in region F′ are LNA nucleosides.
  • the LNA nucleosides in region F′ are independently selected from the group consisting of oxy-LNA, thio-LNA, amino-LNA, cET, and/or ENA, in either the beta-D or alpha-L configurations or combinations thereof.
  • region F′ has at least 1 beta-D-oxy LNA unit, at the 5′ end of the contiguous sequence.
  • the oligonucleotide of the invention n comprises a contiguous nucleotide region which is complementary to the target nucleic acid.
  • the oligonucleotide may further comprise additional nucleotides positioned 5′ and/or 3′ to the contiguous nucleotide region, which are referred to as region D herein.
  • Region D′ and D′′ can be attached to the 5′ end of region F or the 3′ end of region F′, respectively.
  • the D regions (region D′ or D′′) may in some embodiments form part of the contiguous nucleotide sequence which is complementary to the target nucleic acid, or in other embodiments the D region(s) may be non-complementary to the target nucleic acid.
  • the oligonucleotide of the invention consists or comprises of the contiguous nucleotide region and optionally 1-5 additional 5′ nucleotides (region D′).
  • the oligonucleotide of the invention consists or comprises of the contiguous nucleotide region and optionally 1-5 additional 3′ nucleotides (region D′′).
  • Region D′ or D′′ may independently comprise 1, 2, 3, 4 or 5 additional nucleotides, which may be complementary or non-complementary to the target nucleic acid.
  • the oligonucleotide of the invention may in some embodiments comprise a contiguous nucleotide sequence capable of modulating the target which is flanked at the 5′ and/or 3′ end by additional nucleotides.
  • additional nucleotides may serve as a nuclease susceptible biocleavable linker, and may therefore be used to attach a functional group such as a conjugate moiety to the oligonucleotide of the invention.
  • the additional 5′ and/or 3′ end nucleotides are linked with phosphodiester linkages, and may be DNA or RNA.
  • the additional 5′ and/or 3′ end nucleotides are modified nucleotides which may for example be included to enhance nuclease stability or for ease of synthesis.
  • the oligonucleotide of the invention comprises a region D′ and/or D′′ in addition to the contiguous nucleotide region.
  • the gapmer oligonucleotide of the present invention can be represented by the following formulae:
  • F-G-F′ in particular F 1-7 -G 4-12 -F′ 1-7
  • D′-F-G-F′ in particular D′ 1-3 —F 1-7 -G 4-12 -F′ 1-7
  • F-G-F′-D′′ in particular F 1-7 -G 4-12 -F′ 1-7 -D′′ 1-3
  • D′-F-G-F′-D′′ in particular D′ 1-3 —F 1-7 -G 4-12 -F′ 1-7 -D′′ 1-3
  • the invention provides methods for manufacturing the oligonucleotides of the invention comprising reacting nucleotide units and thereby forming covalently linked contiguous nucleotide units comprised in the oligonucleotide.
  • the method uses phophoramidite chemistry (see for example Caruthers et al, 1987, Methods in Enzymology vol. 154, pages 287-313).
  • the method further comprises reacting the contiguous nucleotide sequence with a conjugating moiety (ligand).
  • composition of the invention comprising mixing the oligonucleotide or conjugated oligonucleotide of the invention with a pharmaceutically acceptable diluent, solvent, carrier, salt and/or adjuvant.
  • the oligonucleotide of the invention may be provided as a suitable pharmaceutical salt, such as a sodium or potassium salt.
  • a suitable pharmaceutical salt such as a sodium or potassium salt.
  • the oligonucleotide of the invention is a sodium salt.
  • the invention provides pharmaceutical compositions comprising any of the aforementioned oligonucleotides and/or oligonucleotide conjugates and a pharmaceutically acceptable diluent, carrier, salt and/or adjuvant.
  • a pharmaceutically acceptable diluent includes phosphate-buffered saline (PBS) and pharmaceutically acceptable salts include, but are not limited to, sodium and potassium salts.
  • the pharmaceutically acceptable diluent is sterile phosphate buffered saline.
  • the oligonucleotide is used in the pharmaceutically acceptable diluent at a concentration of 50-300 ⁇ M solution.
  • the oligonucleotide of the invention is administered at a dose of 10-1000 ⁇ g.
  • WO 2007/031091 provides suitable and preferred examples of pharmaceutically acceptable diluents, carriers and adjuvants (hereby incorporated by reference). Suitable dosages, formulations, administration routes, compositions, dosage forms, combinations with other therapeutic agents, pro-drug formulations are also provided in WO2007/031091.
  • Oligonucleotides or oligonucleotide conjugates of the invention may be mixed with pharmaceutically acceptable active or inert substances for the preparation of pharmaceutical compositions or formulations.
  • Compositions and methods for the formulation of pharmaceutical compositions are dependent upon a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered.
  • the oligonucleotide or oligonucleotide conjugate of the invention is a prodrug.
  • the conjugate moiety is cleaved of the oligonucleotide once the prodrug is delivered to the site of action, e.g. the target cell.
  • oligonucleotides of the invention may be utilized as research reagents for, for example, diagnostics, therapeutics and prophylaxis.
  • such oligonucleotides may be used to specifically modulate the synthesis of HTRA1 protein in cells (e.g. in vitro cell cultures) and experimental animals thereby facilitating functional analysis of the target or an appraisal of its usefulness as a target for therapeutic intervention.
  • the target modulation is achieved by degrading or inhibiting the mRNA producing the protein, thereby prevent protein formation or by degrading or inhibiting a modulator of the gene or mRNA producing the protein.
  • the oligonucleotides may be used to detect and quantitate HTRA1 expression in cell and tissues by northern blotting, in-situ hybridisation or similar techniques.
  • an animal or a human suspected of having a disease or disorder, which can be treated by modulating the expression of HTRA1.
  • the invention provides methods for treating or preventing a disease, comprising administering a therapeutically or prophylactically effective amount of an oligonucleotide, an oligonucleotide conjugate or a pharmaceutical composition of the invention to a subject suffering from or susceptible to the disease.
  • the invention also relates to an oligonucleotide, a composition or a conjugate as defined herein for use as a medicament.
  • oligonucleotide, oligonucleotide conjugate or a pharmaceutical composition according to the invention is typically administered in an effective amount.
  • the invention also provides for the use of the oligonucleotide or oligonucleotide conjugate of the invention as described for the manufacture of a medicament for the treatment of a disorder as referred to herein, or for a method of the treatment of as a disorder as referred to herein.
  • the disease or disorder is associated with expression of HTRA1.
  • disease or disorder may be associated with a mutation in the HTRA1 gene or a gene whose protein product is associated with or interacts with HTRA1. Therefore, in some embodiments, the target nucleic acid is a mutated form of the HTRA1 sequence and in other embodiments, the target nucleic acid is a regulator of the HTRA1 sequence.
  • the methods of the invention are preferably employed for treatment or prophylaxis against diseases caused by abnormal levels and/or activity of HTRA1.
  • the invention further relates to use of an oligonucleotide, oligonucleotide conjugate or a pharmaceutical composition as defined herein for the manufacture of a medicament for the treatment of abnormal levels and/or activity of HTRA1.
  • the invention relates to oligonucleotides, oligonucleotide conjugates or pharmaceutical compositions for use in the treatment of diseases or disorders selected from eye disorders, such as macular degeneration, including age related macular degeneration (AMD), such as dry AMD or wet AMD, and diabetic retinopathy.
  • AMD age related macular degeneration
  • the oligonucleotide conjugates or pharmaceutical compositions of the invention may be for use in the treatment of geographic atrophy or intermediate dAMD.
  • HTRA1 has also been indicated in Alzheimer's and Parkinson's disease, and therefore in some embodiments, the oligonucleotide conjugates or pharmaceutical compositions of the invention may be for use in the treatment of Alzheimer's or Parkinson's.
  • HTRA1 has also been indicated in Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, and therefore in some embodiments, the oligonucleotide conjugates or pharmaceutical compositions of the invention may be for use in the treatment of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, or familial ischemic cerebral small-vessel disease.
  • oligonucleotides or pharmaceutical compositions of the present invention may be administered topical (such as, to the skin, inhalation, ophthalmic or otic) or enteral (such as, orally or through the gastrointestinal tract) or parenteral (such as, intravenous, subcutaneous, intra-muscular, intracerebral, intracerebroventricular or intrathecal).
  • topical such as, to the skin, inhalation, ophthalmic or otic
  • enteral such as, orally or through the gastrointestinal tract
  • parenteral such as, intravenous, subcutaneous, intra-muscular, intracerebral, intracerebroventricular or intrathecal.
  • the oligonucleotide, conjugate or pharmaceutical compositions of the present invention are administered by a parenteral route including intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion, intrathecal or intracranial, e.g. intracerebral or intraventricular, administration.
  • the active oligonucleotide or oligonucleotide conjugate is administered intravenously.
  • the active oligonucleotide or oligonucleotide conjugate is administered subcutaneously.
  • intraocular injection may be used.
  • macular degeneration e.g. AMD (wet or dry)
  • intraocular injection may be used.
  • the compound of the invention, or pharmaceutically acceptable salt thereof is administered via an intraocular injection in a dose from about 10 ⁇ g to about 200 ⁇ g per eye, such as about 50 ⁇ g to about 150 ⁇ g per eye, such as about 100 ⁇ g per eye.
  • the dosage interval i.e. the period of time between consecutive dosings is at least monthly, such as at least bi monthly or at least once every three months.
  • the oligonucleotide, oligonucleotide conjugate or pharmaceutical composition of the invention is for use in a combination treatment with another therapeutic agent.
  • the therapeutic agent can for example be the standard of care for the diseases or disorders described above
  • An antisense oligonucleotide of 10-30 nucleotides in length wherein said antisense oligonucleotide comprises a contiguous nucleotide region of 10-22 nucleotides which are at least 90% such as 100% complementarity to SEQ ID NO 147:
  • SEQ ID NO 138 CAAATATTTACCTGGTTG SEQ ID NO 139: TTTACCTGGTTGTTGG SEQ ID NO 140: CCAAATATTTACCTGGTT SEQ ID NO 141: CCAAATATTTACCTGGTTGT SEQ ID NO 142: ATATTTACCTGGTTGTTG SEQ ID NO 143: TATTTACCTGGTTGTT SEQ ID NO 144: ATATTTACCTGGTTGT SEQ ID NO 145: ATATTTACCTGGTTGTT
  • SEQ ID NO 146 TTTACCTGGTT
  • the one or more 2′ sugar modified nucleoside is independently selected from the group consisting of 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA, 2′-amino-DNA, 2′-fluoro-DNA, arabino nucleic acid (ANA), 2′-fluoro-ANA and LNA nucleosides.
  • region G consists or comprises 6-16 DNA nucleosides.
  • region F and F′ each comprise at least one LNA nucleoside.
  • a capital letter represents an LNA nucleoside unit
  • a lower case letter represents a DNA nucleoside unit
  • subscript s represents a phosphorothioate internucleoside linkage, wherein all LNA cytosines are 5-methyl cytosine.
  • a conjugate comprising the oligonucleotide according to any one of embodiments 1-17, and at least one conjugate moiety covalently attached to said oligonucleotide.
  • a pharmaceutical composition comprising the oligonucleotide of embodiment 1-17 or the conjugate of embodiment 18 and a pharmaceutically acceptable diluent, solvent, carrier, salt and/or adjuvant.
  • a method for treating or preventing a disease comprising administering a therapeutically or prophylactically effective amount of an oligonucleotide of any one of embodiments 1-17 or the conjugate according to embodiment 18 or the pharmaceutical composition of embodiment 19 to a subject suffering from or susceptible to the disease.
  • the disease is selected from the group consisting of macular degeneration (such as wetAMD, dryAMD, geographic atrophy, intermediate dAMD, diabetic retinopathy), Parkinson's disease, Alzhiemer's disease, Duchenne muscular dystrophy, arthritis, such as osteoarthritis, and familial ischemic cerebral small-vessel disease.
  • macular degeneration such as wetAMD, dryAMD, geographic atrophy, intermediate dAMD, diabetic retinopathy
  • Parkinson's disease Alzhiemer's disease
  • Duchenne muscular dystrophy arthritis, such as osteoarthritis, and familial ischemic cerebral small-vessel disease.
  • the oligonucleotide of any one of embodiments 1-17 or the conjugate according to embodiment 18 or the pharmaceutical composition of embodiment 19 for use in the treatment or prevention of a disease is selected from the group consisting of macular degeneration (such as wetAMD, dryAMD, geographic atrophy, intermediate dAMD, diabetic retinopathy), Parkinson's disease, Alzhiemer's disease, Duchenne muscular dystrophy, arthritis, such as osteoarthritis, and familial ischemic cerebral small-vessel disease.
  • macular degeneration such as wetAMD, dryAMD, geographic atrophy, intermediate dAMD, diabetic retinopathy
  • Parkinson's disease Alzhiemer's disease
  • Duchenne muscular dystrophy arthritis, such as osteoarthritis, and familial ischemic cerebral small-vessel disease.
  • Use of the oligonucleotide of embodiment 1-17 or the conjugate according to embodiment 18 or the pharmaceutical composition of embodiment 19, for the preparation of a medicament for treatment or prevention of a disease is selected from the group consisting of macular degeneration (such as wetAMD, dryAMD, geographic atrophy, intermediate dAMD, diabetic retinopathy), Parkinson's disease, Alzhiemer's disease, Duchenne muscular dystrophy, arthritis, such as osteoarthritis, and familial ischemic cerebral small-vessel disease.
  • macular degeneration such as wetAMD, dryAMD, geographic atrophy, intermediate dAMD, diabetic retinopathy
  • Parkinson's disease Alzhiemer's disease
  • Duchenne muscular dystrophy arthritis, such as osteoarthritis, and familial ischemic cerebral small-vessel disease.
  • Oligonucleotide synthesis is generally known in the art. Below is a protocol which may be applied. The oligonucleotides of the present invention may have been produced by slightly varying methods in terms of apparatus, support and concentrations used.
  • Oligonucleotides are synthesized on uridine universal supports using the phosphoramidite approach on an Oligomaker 48 at 1 ⁇ mol scale. At the end of the synthesis, the oligonucleotides are cleaved from the solid support using aqueous ammonia for 5-16 hours at 60° C. The oligonucleotides are purified by reverse phase HPLC (RP-HPLC) or by solid phase extractions and characterized by UPLC, and the molecular mass is further confirmed by ESI-MS.
  • RP-HPLC reverse phase HPLC
  • UPLC UPLC
  • the coupling of ⁇ -cyanoethyl-phosphoramidites is performed by using a solution of 0.1 M of the 5′-O-DMT-protected amidite in acetonitrile and DCI (4,5-dicyanoimidazole) in acetonitrile (0.25 M) as activator.
  • a phosphoramidite with desired modifications can be used, e.g. a C6 linker for attaching a conjugate group or a conjugate group as such.
  • Thiolation for introduction of phosphorthioate linkages is carried out by using xanthane hydride (0.01 M in acetonitrile/pyridine 9:1). Phosphordiester linkages can be introduced using 0.02 M iodine in THF/Pyridine/water 7:2:1. The rest of the reagents are the ones typically used for oligonucleotide synthesis.
  • conjugation For post solid phase synthesis conjugation a commercially available C6 aminolinker phorphoramidite can be used in the last cycle of the solid phase synthesis and after deprotection and cleavage from the solid support the aminolinked deprotected oligonucleotide is isolated.
  • the conjugates are introduced via activation of the functional group using standard synthesis methods.
  • the crude compounds are purified by preparative RP-HPLC on a Phenomenex Jupiter C18 10 ⁇ 150 ⁇ 10 mm column. 0.1 M ammonium acetate pH 8 and acetonitrile is used as buffers at a flow rate of 5 mL/min. The collected fractions are lyophilized to give the purified compound typically as a white solid.
  • Oligonucleotide and RNA target (phosphate linked, PO) duplexes are diluted to 3 mM in 500 ml RNase-free water and mixed with 500 ml 2 ⁇ T m -buffer (200 mM NaCl, 0.2 mM EDTA, 20 mM Naphosphate, pH 7.0). The solution is heated to 95° C. for 3 min and then allowed to anneal in room temperature for 30 min.
  • the duplex melting temperatures (T m ) is measured on a Lambda 40 UV/VIS Spectrophotometer equipped with a Peltier temperature programmer PTP6 using PE Templab software (Perkin Elmer). The temperature is ramped up from 20° C. to 95° C. and then down to 25° C., recording absorption at 260 nm. First derivative and the local maximums of both the melting and annealing are used to assess the duplex T m .
  • Rat C6 cell line was purchased from ATCC and maintained as recommended by the supplier in a humidified incubator at 37° C. with 5% CO 2 .
  • 1500 C6 cells/well were seeded in a 96 multi well plate in culture media. Cells were incubated for 2 hours before addition of oligonucleotides dissolved in PBS. Concentration of oligonucleotides: 25 ⁇ M. 4 days after addition of oligonucleotides, the cells were harvested. RNA was extracted using the PureLink Pro 96 RNA Purification kit (Ambion, according to the manufacturer's instructions).
  • cDNA was then synthesized using M-MLT Reverse Transcriptase, random decamers RETROscript, RNase inhibitor (Ambion, according the manufacturer's instruction) with 100 mM dNTP set PCR Grade (Invitrogen) and DNase/RNase free Water (Gibco).
  • qPCR was performed using TagMan Fast Advanced Master Mix (2 ⁇ ) (Ambion) in a doublex set up. Following TaqMan primer assays were used for qPCR: Htra1, Rn00581870_m1 (FAM-MGB) and house keeping gene, Tbp, Rn01455646_m1 (VIC-MGB). All primer sets were purchase from Life Technologies. The relative Htra1 mRNA expression level in the table is shown as % of control (PBS-treated cells).
  • Human glioblastoma U251 cell line was purchased from ECACC and maintained as recommended by the supplier in a humidified incubator at 37° C. with 5% CO 2 .
  • 15000 U251 cells/well were seeded in a 96 multi well plate in starvation media (media recommended by the supplier with the exception of 1% FBS instead of 10%). Cells were incubated for 24 hours before addition of oligonucleotides dissolved in PBS. Concentration of oligonucleotides: 5 ⁇ M. 3-4 days after addition of compounds, media was removed and new media (without oligonucleotide) was added. 6 days after addition of oligonucleotides, the cells were harvested.
  • HTRA1 mRNA expression level in the table is shown as % of control (PBS-treated cells).
  • Human retinal pigmented epithelium ARPE19 cell line was purchased by from ATCC and maintained in DMEM-F12 (Sigma, D8437), 10% FBS, 1% pen/strep in a humidified incubator at 37° C. with 5% CO 2 .
  • the U251 cell line was described in example 3.
  • 5000 ARPE19 cells/well were seeded in a 96 multi well plate in culture media (with the exception of 5% FBS instead of 10%). Cells were incubated for 1 hour before addition of oligonucleotides dissolved in PBS. 4 days after addition of oligonucleotides, the cells were harvested.
  • the assay with the U251 cell line was performed as described in example 3.
  • RNA was extracted using the RNeasy 96 Biorobot 8000 kit (Qiagen, according to the manufacturer's instructions). cDNA was then synthesized using Retroscript cDNA synthesis kit (ThermoFisher, according the manufacturer's instruction). For gene expressions analysis, qPCR was performed using the Fluidigm Biomark system. Following TaqMan primer assays were used for qPCR: HTRA1, Hs01016151_m1 and house-keeping genes, TBP, Hs99999910_m1 and PPIA, Hs99999904_m1, from Life Technologies. n 2 biological replicates. The relative HTRA1 mRNA expression level is shown in the table as % of control (PBS). Additional HTRA1 primer set (Hs00170197_m1) was also tested and the same trends were observed (data not shown).
  • ARPE19, U251 and C6 cell lines were described in example 4, 3 and 1, respectively.
  • 2000 U251 or ARPE19 cells/well were seeded in a 96 multi well plate in culture media recommended by the supplier. Cells were incubated for 2 hours before addition of oligonucleotides dissolved in PBS.
  • the C6 cell line assay was performed as described in example 1-2. Concentration of oligonucleotides: from 50 ⁇ M, half-log dilution, 8 points. 4 days after addition of oligonucleotides, the cells were harvested. RNA extraction, cDNA synthesis and qPCR were performed for all cell lines as described in Example 1.
  • HTRA1 Hs01016151_m1
  • TBP house-keeping gene
  • Hs4326322E VIC-MGB
  • ARPE19 and U251 cell lines and assays were described in example 6.
  • RNA extraction was performed as described in example 1, cDNA synthesis and qPCR were performed using qScript XLT one-step RT-qPCR ToughMix Low ROX, 95134-100 (Quanta Biosciences).
  • the relative HTRA1 mRNA expression level in the table is shown as % of control (PBS-treated cells).
  • hpRPE Human primary Retinal Pigmented Epithelium
  • MEM Alpha media (Sigma Cat #M-4526) supplemented with N1 supplement (Sigma Cat #N-6530), Glutamine-Penicillin-Streptomycin (Sigma Cat #G-1146), Non-Essential Amino Acid (NEAA, Sigma Cat #M-7145), Taurine (Sigma Cat #T-0625), Hydrocortisone (Sigma Cat #H-03966), Triiodo-thyronin (Sigma Cat #T-5516) and Bovine Serum Albumin (BSA, Sigma Cat #A-9647).
  • MEM Alpha media (Sigma Cat #M-4526) supplemented with N1 supplement (Sigma Cat #N-6530), Glutamine-Penicillin-Streptomycin (Sigma Cat #G-1146), Non-Essential Amino Acid (NEAA, Sigma Cat #M-7145), Taurine (Sigma Cat #T-0625), Hydrocortisone (Sigma Cat #H-03966), Triiodo-thyronin (
  • RNA quality control is performed with the Agilent Bioanalyzer Nano Kit (Agilent; Cat #5067-1511; Lot 1446).
  • cDNA synthesis Reverse transcription of total RNA into cDNA (cDNA synthesis) is performed using the High Capacity cDNA Reverse Transcription Kit which is based on random hexamer oligonucleotides, according to the manufacturer's instructions (Thermo Fisher Scientific, Cat #4368814; Lot 00314158). The measurement of the cDNA samples is carried out in triplicates, in a 384-well plate format on the 7900HT real-time PCR instrument (Thermo Fisher Scientific).
  • HTRA1 Hs01016151_m1 and Hs00170197_m1
  • housekeeping genes GAPDH, Hs99999905_m1 and PPIA, Hs99999904_m1 from Life Technologies.
  • n 3 biological replicates.
  • the relative HTRA1 mRNA expression level is shown in the table as % of control (PBS). See FIG. 3 .
  • Example 9 Rat In Vivo Efficacy Study, 7 Days of Treatment, Intravitreal (IVT) Injection, 30 ⁇ g/Eye
  • the animals were anesthetized with isoflurane, eyes were disinfected and dilated before an intravitreal injection of 30 ⁇ g (in 3 ⁇ l) per eye.
  • RNA quality control was performed with the Agilent Bioanalyzer Nano Kit (Agilent; Cat #5067-1511; Lot 1446).
  • RNA into cDNA was performed using the High Capacity cDNA Reverse Transcription Kit which is based on random hexamer oligonucleotides, according to the manufacturer's instructions (Thermo Fisher Scientific, Cat #4368814, Lot 00314158).
  • the measurement of the cDNA samples was carried out in triplicates, in a 384-well plate format on the 7900HT real-time PCR instrument (Thermo Fisher Scientific).
  • the relative Htra1 mRNA expression level is shown as % of control (PBS). See FIG. 4 .
  • the animals were anesthetized with an intramuscular injection of a mix of xylazine and ketamine.
  • the test item and negative control (PBS) were administered intravitreally in both eyes of anesthetized animals (3 ⁇ L per administration) on study day 1.
  • the samples were diluted 10-50 fold for oligo content measurements with a hybridization ELISA method.
  • a biotinylated LNA-capture probe and a digoxigenin-conjugated LNA-detection probe (both 35 nM in 5 ⁇ SSCT, each complementary to one end of the LNA oligonucleotide to be detected) was mixed with the diluted homogenates or relevant standards, incubated for 30 minutes at RT and then added to a streptavidine-coated ELISA plates (Nunc cat. no. 436014).
  • the plates were incubated for 1 hour at RT, washed in 2 ⁇ SSCT (300 mM sodium chloride, 30 mM sodium citrate and 0.05% v/v Tween-20, pH 7.0)
  • the captured LNA duplexes were detected using an anti-DIG antibodies conjugated with alkaline phosphatase (Roche Applied Science cat. No. 11093274910) and an alkaline phosphatase substrate system (Blue Phos substrate, KPL product code 50-88-00).
  • the amount of oligo complexes was measured as absorbance at 615 nm on a Biotek reader.
  • RNA extraction For RNA extraction, cellular RNA large volume kit (05467535001, Roche) was used in the MagNA Pure 96 system with the program: Tissue FF standard LV3.1 according to the instructions of the manufacturer, including DNAse treatment. RNA quality control and concentration were measured with an Eon reader (Biotek). The RNA concentration was normalized across samples, and subsequent cDNA synthesis and qPCR was performed in a one-step reaction using qScript XLT one-step RT-qPCR ToughMix Low ROX, 95134-100 (Quanta Biosciences).
  • TaqMan primer assays were used in a duplex reaction: Htra1, Rn00581870_m1 and Rn00668987_m1 and housekeeping genes, HPRT, Rn01527840_m1 and Tbp, Rn01455646_m1, from Life Technologies.
  • the animals were anesthetized with an intramuscular injection of a mix of xylazine and ketamine.
  • the test item and negative control (PBS) were administered intravitreally in both eyes of anesthetized animals (3 ⁇ L per administration) on study day ⁇ 3.
  • the positive control item was injected intraperitoneally on Day 0, 4 times (0.5 h before starting light exposure, 2 h and 4 h after starting light exposure and just after the end of light exposure), at a dose volume of 2.5 mL/kg, using a 25-gauge needle mounted on a 1 mL-plastic syringe, protected from light.
  • the rats were dark adapted for 36 hours and then exposed to a continuous blue fluorescent light (400-540 nm) in clear plastic cages for 6 hours. After exposure, the rats were placed in dark room for 24 hours before returning to standard cyclic light conditions.
  • a continuous blue fluorescent light 400-540 nm
  • Electroretinogram (ERG) Electroretinogram
  • Electroretinograms were be recorded at baseline and on Day 14 on both eyes after overnight darkadaption. A-wave and b-wave amplitudes were measured for each ERG recording
  • V Vitreous
  • R Retina
  • CH Choroid
  • the samples were diluted 10-50 fold for oligo content measurements with a hybridization ELISA method.
  • a biotinylated LNA-capture probe and a digoxigenin-conjugated LNA-detection probe (both 35 nM in 5 ⁇ SSCT, each complementary to one end of the LNA oligonucleotide to be detected) was mixed with the diluted homogenates or relevant standards, incubated for 30 minutes at RT and then added to a streptavidine-coated ELISA plates (Nunc cat. no. 436014).
  • the plates were incubated for 1 hour at RT, washed in 2 ⁇ SSCT (300 mM sodium chloride, 30 mM sodium citrate and 0.05% v/v Tween-20, pH 7.0)
  • the captured LNA duplexes were detected using an anti-DIG antibodies conjugated with alkaline phosphatase (Roche Applied Science cat. No. 11093274910) and an alkaline phosphatase substrate system (Blue Phos substrate, KPL product code 50-88-00).
  • the amount of oligo complexes was measured as absorbance at 615 nm on a Biotek reader.
  • RNA extraction For RNA extraction, cellular RNA large volume kit (05467535001, Roche) was used in the MagNA Pure 96 system with the program: Tissue FF standard LV3.1 according to the instructions of the manufacturer, including DNAse treatment. RNA quality control and concentration were measured with an Eon reader (Biotek). The RNA concentration was normalized across samples, and subsequent cDNA synthesis and qPCR was performed in a one-step reaction using qScript XLT one-step RT-qPCR ToughMix Low ROX, 95134-100 (Quanta Biosciences).
  • TaqMan primer assays were used in a duplex reaction: Htra1, Rn00581870_m1 and Rn00668987_m1 and housekeeping genes, HPRT, Rn01527840_m1 and Tbp, Rn01455646_m1, from Life Technologies.
  • ISH RNAscope was performed as described in example 10.
  • Knockdown (KD) at mRNA level was observed in the retina for 2 selected HTRA1 LNA oligonucleotides targeting the “hotspot” in human HTRA1 pre-mRNA between position 33042-33064 (SEQ ID NO 147). This was observed both with qPCR and ISH readouts (see FIGS. 7A and /B and the following table).
  • the variation of the knockdown is relatively large, see the standard deviations listed in the table. The variation seems to be at the administration level which can be seen when plotting a dose response curve for oligo content vs. residual HTRA1 mRNA level (see FIG. 7C ).
  • the animals were anesthetized in isofluran.
  • the test item and negative control (PBS) were administered intravitreally in both eyes of anesthetized animals (3 ⁇ L per administration) on study day 1.
  • the relative residual Htra1 mRNA expression level is shown as % of control (PBS).
  • Buprenorphine analgesia was administered prior to, and two days after test compound injection.
  • the animals were anesthetized with an intramuscular injection of ketamine and xylazine.
  • the test item and negative control (PBS) were administered intravitreally in both eyes of anesthetized animals (50 ⁇ L per administration) on study day 1 after local application of tetracaine anesthetic.
  • the samples were diluted 10-50 fold for oligo content measurements with a hybridization ELISA method.
  • a biotinylated LNA-capture probe and a digoxigenin-conjugated LNA-detection probe (both 35 nM in 5 ⁇ SSCT, each complementary to one end of the LNA oligonucleotide to be detected) was mixed with the diluted homogenates or relevant standards, incubated for 30 minutes at RT and then added to a streptavidine-coated ELISA plates (Nunc cat. no. 436014).
  • the plates were incubated for 1 hour at RT, washed in 2 ⁇ SSCT (300 mM sodium chloride, 30 mM sodium citrate and 0.05% v/v Tween-20, pH 7.0)
  • the captured LNA duplexes were detected using an anti-DIG antibodies conjugated with alkaline phosphatase (Roche Applied Science cat. No. 11093274910) and an alkaline phosphatase substrate system (Blue Phos substrate, KPL product code 50-88-00).
  • the amount of oligo complexes was measured as absorbance at 615 nm on a Biotek reader.
  • RNA extraction For RNA extraction, cellular RNA large volume kit (05467535001, Roche) was used in the MagNA Pure 96 system with the program: Tissue FF standard LV3.1 according to the instructions of the manufacturer, including DNAse treatment. RNA quality control and concentration were measured with an Eon reader (Biotek). The RNA concentration was normalized across samples, and subsequent cDNA synthesis and qPCR was performed in a one-step reaction using qScript XLT one-step RT-qPCR ToughMix Low ROX, 95134-100 (Quanta Biosciences).
  • TaqMan primer assays were used in singplex reactions: Htra1, Mf01016150_, Mf01016152_m1 and Rh02799527_m1 and housekeeping genes, ARFGAP2, Mf01058488_g1 and Rh01058485_m1, and ARL1, Mf02795431_m1, from Life Technologies.
  • Eyeballs were removed and fixed in 10% neutral buffered formalin for 24 hours, trimmed and embedded in paraffin.
  • ISH analysis sections of formalin-fixed, paraffin-embedded retina tissue 4 ⁇ m thick were processed using the fully automated Ventana Dicovery ULTRA Staining Module (Procedure: mRNA Discovery Ultra Red 4.0-v0.00.0152) using the RNAscope 2.5 VS Probe-Mmu-HTRA1, REF 486979, Advanced Cell Diagnostics, Inc. Chromogen used is Fastred, Hematoxylin H counterstain.
  • IP-MS Plate-Based Immunoprecipitation Mass Spectrometry
  • Retinas were homogenized in 4 volumes (w/v) of RIPA buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 0.25% deoxycholic acid, 1% NP-40, 1 mM EDTA, Millipore) with protease inhibitors (Complete EDTA-free, Roche) using a Precellys 24 (5500, 15 s, 2 cycles). Homogenates were centrifuged (13,000 rpm, 3 min) and the protein contents of the supernatants determined (Pierce BCA protein assay)
  • Vitreous humors (300 ⁇ l) were diluted with 5 ⁇ RIPA buffer (final concentration: 50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 0.25% deoxycholic acid, 1% NP-40, 1 mM EDTA) with protease inhibitors (Complete EDTA-free, Roche) and homogenized using a Precellys 24 (5500, 15 s, 2 cycles). Homogenates were centrifuged (13,000 rpm, 3 min) and the protein contents of the supernatants determined (Pierce BCA protein assay)
  • a 96 well plate (Nunc MaxiSorp) was coated with anti-HTRA1 mouse monoclonal antibody (R&D MAB2916, 500 ng/well in 50 ⁇ l PBS) and incubated overnight at 4° C. The plate was washed twice with PBS (200 ⁇ l) and blocked with 3% (w/v) BSA in PBS for 30 min at 20° C. followed by two PBS washes. Samples (75 ⁇ g retina, 100 ⁇ g vitreous in 50 ⁇ l PBS) were randomized and added to the plate followed by overnight incubation at 4° C. on a shaker (150 rpm). The plate was then washed twice with PBS and once with water.
  • R&D MAB2916 anti-HTRA1 mouse monoclonal antibody
  • Mass spectrometry analysis was performed on an Ultimate RSLCnano LC coupled to a TSQ Quantiva triple quadrupole mass spectrometer (Thermo Scientific). Samples (20 ⁇ L) were injected directly from the 96 well plate used for IP and loaded at 5 ⁇ L/min for 6 min onto a Acclaim Pepmap 100 trap column (100 ⁇ m ⁇ 2 cm, C18, 5 ⁇ m, 100 ⁇ , Thermo Scientific) in loading buffer (0.5% v/v formic acid, 2% v/v ACN).
  • Peptides were then resolved on a PepMap Easy-SPRAY analytical column (75 ⁇ m ⁇ 15 cm, 3 ⁇ m, 100 ⁇ , Thermo Scientific) with integrated electrospray emitter heated to 40° C. using the following gradient at a flow rate of 250 nL/min: 6 min, 98% buffer A (2% ACN, 0.1% formic acid), 2% buffer B (ACN+0.1% formic acid); 36 min, 30% buffer B; 41 min, 60% buffer B; 43 min, 80% buffer B; 49 min, 80% buffer B; 50 min, 2% buffer B.
  • the TSQ Quantiva was operated in SRM mode with the following parameters: cycle time, 1.5 s; spray voltage, 1800 V; collision gas pressure, 2 mTorr; Q1 and Q3 resolution, 0.7 FWHM; ion transfer tube temperature 300° C.
  • SRM transitions were acquired for the HTRA1 peptide “LHRPPVIVLQR” and an isotope labelled (L-[U-13C, U-15N]R) synthetic version, which was used an internal standard.
  • Data analysis was performed using Skyline version 3.6

Abstract

The present invention relates to antisense oligonucleotides (oligomers) that are complementary to HTRA1, leading to modulation of the expression of HTRA1. Modulation of HTRA1 expression is beneficial for a range of medical disorders, such as macular degeneration, e.g. age-related macular degeneration.

Description

    FIELD OF INVENTION
  • The present invention relates to antisense oligonucleotides (oligomers) that are complementary to HTRA1, leading to modulation of the expression of HTRA1. Modulation of HTRA1 expression is beneficial for a range of medical disorders, such as macular degeneration, e.g. age-related macular degeneration.
  • BACKGROUND
  • The human high temperature requirement A (HTRA) family of serine proteases are ubiquitously expressed PDZ-proteases that are involved in maintaining protein homeostasis in extracellular compartments by combining the dual functions of a protease and a chaperone. HTRA housekeeping proteases are implicated in organization of the extracellular matrix, cell proliferation and ageing. Modulation of HTRA activity is connected with severe diseases, including Duchenne muscular dystrophy (Bakay et al. 2002, Neuromuscul. Disord. 12: 125-141), arthritis, such as osteoarthritis (Grau et al. 2006, JBC 281: 6124-6129); cancer, familial ischemic cerebral small-vessel disease and age-related macular degeneration, as well as Parkinson's disease and Alzheimer's disease. The human HTRA1 contains an insulin-like growth factor (IGF) binding domain. It has been proposed to regulate IGF availability and cell growth (Zumbrunn and Trueb, 1996, FEES Letters 398:189-192) and to exhibit tumor suppressor properties. HTRA1 expression is down-regulated in metastatic melanoma, and may thus indicate the degree of melanoma progression. Overexpression of HTRA1 in a metastatic melanoma cell line reduced proliferation and invasion in vitro, and reduced tumor growth in a xenograft mouse model (Baldi et al., 2002, Oncogene 21:6684-6688). HTRA1 expression is also down-regulated in ovarian cancer. In ovarian cancer cell lines, HTRA1 overexpression induces cell death, while antisense HTRA1 expression promoted anchorage-independent growth (Chien et al., 2004, Oncogene 23:1636-1644).
  • In addition to its effect on the IGF pathway, HTRA1 also inhibits signaling by the TGFβ family of growth factors (Oka et al., 2004, Development 131:1041-1053). HTRA1 can cleave amyloid precursor protein (APP), and HTRA1 inhibitors cause the accumulation of Aβ peptide in cultured cells. Thus, HTRA1 is also implicated in Alzheimer's disease (Grau et al., 2005, Proc. Nat. Acad. Sci. USA. 102:6021-6026).
  • On the other hand HTRA1 upregulation has been observed and seems to be associated to Duchenne muscular dystrophy (Bakay et al. 2002, Neuromuscul. Disord. 12: 125-141) and osteoarthritis (Grau et al. 2006, JBC 281: 6124-6129) and AMD (Fritsche, et al. Nat Gen 2013 45(4):433-9.)
  • A single nucleotide polymorphism (SNP) in the HTRA1 promoter region (rs11200638) is associated with a 10 fold increased the risk of developing age-related macular degeneration (AMD). Moreover the HTRA1 SNPs are in linkage disequilibrium with the ARMS2 SNP (rs10490924) associated with increased risk of developing age-related macular degeneration (AMD). The risk allele is associated with 2-3 fold increased HTRA1 mRNA and protein expression, and HTRA1 is present in drusen in patients with AMD (Dewan et al., 2006, Science 314:989-992; Yang et al., 2006, Science 314:992-993). Different animal models have confirmed that over-expression of HtrA1 Induces AMD-like phenotype in mice. The hHTRA transgenic mouse (Veierkottn, PlosOne 2011) reveals degradation of the elastic lamina of Bruch's membrane, determines choroidal vascular abnormalities (Jones, PNAS 2011) and increases the Polypoidal choroidal vasculopathy (PCV) lesions (Kumar, IOVS 2014). Additionally it has been reported Bruch's membrane damage in hHTRA1 Tg mice, which determines upon exposure to cigarette smoke 3 fold increases CNV (Nakayama, IOVS 2014)
  • Age-related macular degeneration (AMD) is the leading cause of irreversible loss of vision in people over the age of 65. With onset of AMD there is gradual loss of the light sensitive photoreceptor cells in the back of the eye, the underlying pigment epithelial cells that support them metabolically, and the sharp central vision they provide. Age is the major risk factor for the onset of AMD: the likelihood of developing AMD triples after age 55. Smoking, light iris color, gender (women are at greater risk), obesity, and repeated exposure to UV radiation also increase the risk of AMD. There are two forms of AMD: dry AMD and wet AMD. In dry AMD, drusen appear in the macula of the eye, the cells in the macula die, and vision becomes blurry. Dry AMD can progress in three stages: 1) early, 2) intermediate, and 3) advanced dry AMD. Dry AMD can also progress into wet AMD during any of these stages. Wet AMD (also known as exudative AMD), is associated with pathologic posterior segment neovascularization. The posterior segment neovascularization (PSNV) found in exudative AMD is characterized as pathologic choroidal neovascularization. Leakage from abnormal blood vessels forming in this process damages the macula and impairs vision, eventually leading to blindness. Treatment strategies for wet AMD are few and palliative at best. There is therefore an unmet medical need in the provision of effective drugs to treat macular degenerative conditions such as wet and dry AMD. WO 2008/013893 claims a composition for treating a subject suffering from age related macular degeneration comprising a nucleic acid molecules comprising an antisense sequence that hybridizes to HTRA1 gene or mRNA: No antisense molecules are disclosed. WO2009/006460 provides siRNAs targeting HTRA1 and their use in treating AMD.
  • Objective of the Invention
  • The present invention provides antisense oligonucleotides which modulate HTRA1 in vivo or in vitro. The invention identified cryptic target sequence motifs present in the human HTRA1 mRNA (including pre-mRNA) which may be targeted by antisense oligonucleotides to give effective HTRA1 inhibition. The invention also provides effective antisense oligonucleotide sequences and compounds which are capable of inhibiting HTRA1, and their use in treatment of diseases or disorders where HTRA1 is indicated.
  • SUMMARY OF INVENTION
  • The present invention relates to oligonucleotides targeting a mammalian HTRA1 nucleic acid, i.e. are capable of inhibiting the expression of HTRA1 and to treat or prevent diseases related to the functioning of the HTRA1. The oligonucleotides targeting HTRA1 are antisense oligonucleotides, i.e. are complementary to their HTRA1 nucleic acid target.
  • The oligonucleotide of the invention may be in the form of a pharmaceutically acceptable salt, such as a sodium salt or a potassium salt.
  • Accordingly, the invention provides antisense oligonucleotides which comprise a contiguous nucleotide sequence of 10-30 nucleotides in length with at least 90% complementarity, such as fully complementary to a mammalian HTRA1 nucleic acid, such as SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 3 or SEQ ID NO 4.
  • In a further aspect, the invention provides pharmaceutical compositions comprising the oligonucleotides of the invention and pharmaceutically acceptable diluents, carriers, salts and/or adjuvants.
  • The invention provides LNA antisense oligonucleotides, such as LNA gapmer oligonucleotides, which comprise a contiguous nucleotide sequence of 10-30 nucleotides in length with at least 90% complementarity, such as fully complementary to a HTRA1 nucleic acid, such as a sequence selected from the group consisting of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 3 or SEQ ID NO 4.
  • The invention provides for an antisense oligonucleotide comprising a contiguous nucleotide region of 10-22, such as 12-22 nucleotides which are at least 90% such as 100% complementarity to SEQ ID NO 147:
  • SEQ ID NO 147: 5′ CCAACAACCAGGTAAATATTTG 3′
  • The invention provides for an antisense oligonucleotide comprising a contiguous nucleotide region of 10-17, such as 11, 12, 13, 14, 15, 16, such as 12-16 or 12-17 nucleotides which are complementarity to a sequence selected from the group consisting of SEQ ID NO 148-155.
  • The invention provides for an antisense oligonucleotide comprising a contiguous nucleotide region of 10-17, such as 11, 12, 13, 14, 15, 16, such as 12-16 or 12-17 nucleotides which are complementarity to SEQ ID NO 148 or 155.
  • The invention provides for an antisense oligonucleotide of 10-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of 10-22 nucleotides which are at least 90% such as 100% complementarity to SEQ ID NO 147:
  • SEQ ID NO 147: 5′ CCAACAACCAGGTAAATATTTG 3′
  • The invention provides for an antisense oligonucleotide of 10-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of at least 10, such as at least 12 contiguous nucleotides which are complementary to a sequence present in a sequence selected from SEQ ID NO 148-155.
  • The invention provides for an antisense oligonucleotide of at least 12 nucleotides in length, wherein said antisense oligonucleotide comprises the contiguous sequence of SEQ ID NO 146
  • SEQ ID NO 146: 5′ TTTACCTGGTT 3′.
  • The invention provides for the oligonucleotides provided in the examples. The invention provides for the oligonucleotide, such as an antisense oligonucleotide, which comprises at least 10, such as at least 12, present in a sequence selected from the group consisting of SEQ ID NO 5-145.
  • The invention provides for a conjugate comprising the oligonucleotide according to the invention, and at least one conjugate moiety covalently attached to said oligonucleotide.
  • The invention provides for a pharmaceutically acceptable salt of the oligonucleotide or conjugate of the invention.
  • In a further aspect, the invention provides methods for in vivo or in vitro method for modulation of HTRA1 expression in a cell which is expressing HTRA1, by administering an oligonucleotide, conjugate or composition of the invention in an effective amount to said cell.
  • In a further aspect the invention provides methods for treating or preventing a disease, disorder or dysfunction associated with in vivo activity of HTRA1 comprising administering a therapeutically or prophylactically effective amount of the oligonucleotide of the invention, or conjugate thereof, to a subject suffering from or susceptible to the disease, disorder or dysfunction.
  • In a further aspect the oligonucleotide or composition of the invention is used for the treatment or prevention of macular degeneration, and other disorders where HTRA1 is implicated.
  • The invention provides for the oligonucleotide or conjugate of the invention, for use in the treatment of a disease or disorder selected from the list comprising of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, Alzhiemer's disease and Parkinson's disease.
  • The invention provides for the oligonucleotide or conjugate of the invention, for use in the treatment of macular degeneration, such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy.
  • The invention provides for the use of the oligonucleotide, conjugate or composition of the invention, for the manufacture of a medicament for the treatment of macular degeneration, such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy.
  • The invention provides for the use of the oligonucleotide, conjugate or composition of the invention, for the manufacture of a medicament for the treatment of a disease or disorder selected from the group consisting of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, Alzhiemer's disease and Parkinson's disease.
  • The invention provides for a method of treatment of a subject suffering from a disease or disorder selected from the group consisting of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, Alzhiemer's disease and Parkinson's disease, said method comprising the step of administering an effective amount of the oligonucleotide, conjugate or composition of the invention to the subject.
  • The invention provides for a method of treatment of a subject suffering from an ocular disease, such as macular degeneration, such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy, said method comprising the step of administering an effective amount of the oligonucleotide, conjugate or composition of the invention to the subject.
  • The invention provides for a method of treatment of a subject suffering from an ocular disease, such as macular degeneration, such as wet or dry age related macular degeneration (e.g. wAMD, dAMD, geographic atrophy, intermediate dAMD) or diabetic retinopathy, said method comprising administering at least two dosages of the oligonucleotide of the invention, or pharmaceutically acceptable salt thereof, in an intraocular injection in a dosage of from about 10 μg-200 μg, wherein the dosage interval between administration consecutive is at least 4 weeks or at least monthly.
  • BRIEF DESCRIPTION OF FIGURES
  • FIG. 1. A library of n=129 HTRA1 LNA oligonucleotides were screened in U251 and ARPE19 cell lines at 25 μM. Read out: HTRA1 qPCR. n=6 oligos located between position 33042-33064 were relatively active.
  • FIG. 2. A library of n=116 HTRA1 LNA oligonucleotides in the 33042-33064 hot spot were screened in U251 and ARPE19 cell lines at 5 and 25 μM, respectively. n=7 oligos were selected for further analysis. Read out: HTRA1 qPCR.
  • FIG. 3. Dose response of HTRA1 mRNA level upon treatment of human primary RPE cells with LNA oligonucleotide 139.1 and 143.1.
  • FIG. 4. Results of analysis of in vivo expression of HTRA1.
  • FIGS. 5A-5D. Rat in vivo efficacy study, 7 days of treatment, IVT administration, dose response. Retina samples from rat eyes treated with PBS, 140.1 or 143.1 were analyzed. Htra1 ISH RNAscope was performed, in A) representative samples and in B) an overview table of results are shown. GFAP IHC was also performed, GFAP is a marker for reactive gliosis and reticular fibrosis. C) Retina samples subjected to Htra1 qPCR. RE: right eye, LE: left eye. D) Oligo content bioanalysis was performed and dose response curves for bioanalysis plotted versus relative mRNA expression is shown. EC50 determinations was made in Graph Pad Prims. For PBS treated samples, the oligo content were set to 0.01 μg/g tissue.
  • FIGS. 6A-6D. Poc study, Blue light-induced retinal degeneration in albino rats A) Recovery of electroretinogram a-wave and b-wave amplitudes 14 days after blue light exposure (%). The bars indicate group means and 95% Cl. Each data point indicates mean of right and left eye values for each study animal. B) ISH RNA scope, examples from 2 different areas of the retina. C) Htra1 qPCR of retina samples. D) PK PD correlation.
  • FIGS. 7A-7C. Rat in vivo efficacy kinetic study, IVT administration, 50 μg/eye, 3, 7, 14 days of treatment. A) HTRA1 mRNA level measured in the retina by qPCR. B) HTRA1 mRNA level quantified by ISH. The residual HTRA1 mRNA expression level is shown as % of control (PBS-treated cells) in A and B & C) Dose response curves of oligo content vs. qPCR data for individual time points.
  • FIGS. 8A-8D. Non Human Primate (NHP) PK/PD study, IVT administration, 25 μg/eye. A) HTRA1 mRNA level measured in the retina by qPCR. B) HTRA1 mRNA level illustrated by ISH. C-D) Quantification of HTRA1 protein level in retina and vitreous, respectively, by IP-MS. Dots show data for individual animals. Error bars show standard deviations for technical replicates (n=3).
  • FIG. 9. A Compound of the invention (Compound ID NO 143.1). The compound may be in the form of a pharmaceutical salt, such as a sodium salt or a potassium salt.
  • FIG. 10. A Compound of the invention (Compound ID No 145.3). The compound may be in the form of a pharmaceutical salt, such as a sodium salt or a potassium salt.
  • FIG. 11. An example of a pharmaceutical salt of compound 143.1. M+ is a suitable cation, typically a positive metal ion, such as a sodium or potassium ion. The stoichiometric ratio of the cation to the oligonucleotide anion will depend on the charge of the cation used. Suitably, cations with one, two or three positive charge (M+, M++, or M+++, may be used). For illustrative purpose, twice as many single+charged cations (monovalent), such as Na+ or K+ are needed as compared to a divalent cation such as Ca2+.
  • FIG. 12. An example of a pharmaceutical salt of compound 145.3. See the figure legend for FIG. 11 for the description of the cation M.
  • DEFINITIONS
  • Oligonucleotide
  • The term “oligonucleotide” as used herein is defined as it is generally understood by the skilled person as a molecule comprising two or more covalently linked nucleosides. Such covalently bound nucleosides may also be referred to as nucleic acid molecules or oligomers. Oligonucleotides are commonly made in the laboratory by solid-phase chemical synthesis followed by purification. When referring to a sequence of the oligonucleotide, reference is made to the sequence or order of nucleobase moieties, or modifications thereof, of the covalently linked nucleotides or nucleosides. The oligonucleotide of the invention is man-made, and is chemically synthesized, and is typically purified or isolated. The oligonucleotide of the invention may comprise one or more modified nucleosides or nucleotides.
  • Antisense Oligonucleotides
  • The term “Antisense oligonucleotide” as used herein is defined as oligonucleotides capable of modulating expression of a target gene by hybridizing to a target nucleic acid, in particular to a contiguous sequence on a target nucleic acid. The antisense oligonucleotides are not essentially double stranded and are therefore not siRNAs. Preferably, the antisense oligonucleotides of the present invention are single stranded.
  • Contiguous Nucleotide Region
  • The term “contiguous nucleotide region” refers to the region of the oligonucleotide which is complementary to the target nucleic acid. The term may be used interchangeably herein with the term “contiguous nucleotide sequence” or “contiguous nucleobase sequence” and the term “oligonucleotide motif sequence”. In some embodiments all the nucleotides of the oligonucleotide are present in the contiguous nucleotide region. In some embodiments the oligonucleotide comprises the contiguous nucleotide region and may, optionally comprise further nucleotide(s), for example a nucleotide linker region which may be used to attach a functional group to the contiguous nucleotide sequence. The nucleotide linker region may or may not be complementary to the target nucleic acid. In some embodiments the internucleoside linkages present between the nucleotides of the contiguous nucleotide region are all phosphorothioate internucleoside linkages. In some embodiments, the contiguous nucleotide region comprises one or more sugar modified nucleosides.
  • Nucleotides
  • Nucleotides are the building blocks of oligonucleotides and polynucleotides, and for the purposes of the present invention include both naturally occurring and non-naturally occurring nucleotides. In nature, nucleotides, such as DNA and RNA nucleotides comprise a ribose sugar moiety, a nucleobase moiety and one or more phosphate groups (which is absent in nucleosides). Nucleosides and nucleotides may also interchangeably be referred to as “units” or “monomers”.
  • Modified Nucleoside
  • The term “modified nucleoside” or “nucleoside modification” as used herein refers to nucleosides modified as compared to the equivalent DNA or RNA nucleoside by the introduction of one or more modifications of the sugar moiety or the (nucleo)base moiety. In a preferred embodiment the modified nucleoside comprise a modified sugar moiety. The term modified nucleoside may also be used herein interchangeably with the term “nucleoside analogue” or modified “units” or modified “monomers”.
  • Modified Internucleoside Linkage
  • The term “modified internucleoside linkage” is defined as generally understood by the skilled person as linkages other than phosphodiester (PO) linkages, that covalently couples two nucleosides together. Nucleotides with modified internucleoside linkage are also termed “modified nucleotides”. In some embodiments, the modified internucleoside linkage increases the nuclease resistance of the oligonucleotide compared to a phosphodiester linkage. For naturally occurring oligonucleotides, the internucleoside linkage includes phosphate groups creating a phosphodiester bond between adjacent nucleosides. Modified internucleoside linkages are particularly useful in stabilizing oligonucleotides for in vivo use, and may serve to protect against nuclease cleavage at regions of DNA or RNA nucleosides in the oligonucleotide of the invention, for example within the gap region of a gapmer oligonucleotide, as well as in regions of modified nucleosides.
  • In an embodiment, the oligonucleotide comprises one or more internucleoside linkages modified from the natural phosphodiester to a linkage that is for example more resistant to nuclease attack. Nuclease resistance may be determined by incubating the oligonucleotide in blood serum or by using a nuclease resistance assay (e.g. snake venom phosphodiesterase (SVPD)), both are well known in the art. Internucleoside linkages which are capable of enhancing the nuclease resistance of an oligonucleotide are referred to as nuclease resistant internucleoside linkages. In some embodiments all of the internucleoside linkages of the oligonucleotide, or contiguous nucleotide sequence thereof, are modified. It will be recognized that, in some embodiments the nucleosides which link the oligonucleotide of the invention to a non-nucleotide functional group, such as a conjugate, may be phosphodiester. In some embodiments all of the internucleoside linkages of the oligonucleotide, or contiguous nucleotide sequence thereof, are nuclease resistant internucleoside linkages.
  • In some embodiments the modified internucleoside linkages may be phosphorothioate internucleoside linkages. In some embodiments, the modified internucleoside linkages are compatible with the RNaseH recruitment of the oligonucleotide of the invention, for example phosphorothioate.
  • In some embodiments the internucleoside linkage comprises sulphur (S), such as a phosphorothioate internucleoside linkage.
  • A phosphorothioate internucleoside linkage is particularly useful due to nuclease resistance, beneficial pharmacokinetics and ease of manufacture. In some embodiments all of the internucleoside linkages of the oligonucleotide, or contiguous nucleotide sequence thereof, are phosphorothioate.
  • Nucleobase
  • The term nucleobase includes the purine (e.g. adenine and guanine) and pyrimidine (e.g. uracil, thymine and cytosine) moiety present in nucleosides and nucleotides which form hydrogen bonds in nucleic acid hybridization. In the context of the present invention the term nucleobase also encompasses modified nucleobases which may differ from naturally occurring nucleobases, but are functional during nucleic acid hybridization. In this context “nucleobase” refers to both naturally occurring nucleobases such as adenine, guanine, cytosine, thymidine, uracil, xanthine and hypoxanthine, as well as non-naturally occurring variants. Such variants are for example described in Hirao et al (2012) Accounts of Chemical Research vol 45 page 2055 and Bergstrom (2009) Current Protocols in Nucleic Acid Chemistry Suppl. 37 1.4.1.
  • In a some embodiments the nucleobase moiety is modified by changing the purine or pyrimidine into a modified purine or pyrimidine, such as substituted purine or substituted pyrimidine, such as a nucleobased selected from isocytosine, pseudoisocytosine, 5-methyl cytosine, 5-thiozolo-cytosine, 5-propynyl-cytosine, 5-propynyl-uracil, 5-bromouracil 5-thiazolo-uracil, 2-thio-uracil, 2′thio-thymine, inosine, diaminopurine, 6-aminopurine, 2-aminopurine, 2,6-diaminopurine and 2-chloro-6-aminopurine.
  • The nucleobase moieties may be indicated by the letter code for each corresponding nucleobase, e.g. A, T, G, C or U, wherein each letter may optionally include modified nucleobases of equivalent function. For example, in the exemplified oligonucleotides, the nucleobase moieties are selected from A, T, G, C, and 5-methyl cytosine. Optionally, for LNA gapmers, 5-methyl cytosine LNA nucleosides may be used. In some embodiments, the cytosine nucleobases in a 5′cg3′ motif is 5-methyl cytosine.
  • Modified Oligonucleotide
  • The term modified oligonucleotide describes an oligonucleotide comprising one or more sugar-modified nucleosides and/or modified internucleoside linkages. The term chimeric” oligonucleotide is a term that has been used in the literature to describe oligonucleotides with modified nucleosides.
  • Complementarity
  • The term complementarity describes the capacity for Watson-Crick base-pairing of nucleosides/nucleotides. Watson-Crick base pairs are guanine (G)—cytosine (C) and adenine (A)—thymine (T)/uracil (U). It will be understood that oligonucleotides may comprise nucleosides with modified nucleobases, for example 5-methyl cytosine is often used in place of cytosine, and as such the term complementarity encompasses Watson Crick base-paring between non-modified and modified nucleobases (see for example Hirao et al (2012) Accounts of Chemical Research vol 45 page 2055 and Bergstrom (2009) Current Protocols in Nucleic Acid Chemistry Suppl. 37 1.4.1).
  • The term “% complementary” as used herein, refers to the number of nucleotides in percent of a contiguous nucleotide region or sequence in a nucleic acid molecule (e.g. oligonucleotide) which, at a given position, are complementary to (i.e. form Watson Crick base pairs with) a contiguous nucleotide sequence, at a given position of a separate nucleic acid molecule (e.g. the target nucleic acid). The percentage is calculated by counting the number of aligned bases that form pairs between the two sequences, dividing by the total number of nucleotides in the oligonucleotide and multiplying by 100. In such a comparison a nucleobase/nucleotide which does not align (form a base pair) is termed a mismatch.
  • It will be understood that when referring to complementarity between two sequences, the determination of complementarity is measured across the length of the shorter of the two sequences, such as the length of the contiguous nucleotide region or sequence.
  • The term “fully complementary”, refers to 100% complementarity. In the absence of a % term value or indication of a mismatch, complementary means fully complementary.
  • Identity
  • The term “Identity” as used herein, refers to the number of nucleotides in percent of a contiguous nucleotide sequence in a nucleic acid molecule (e.g. oligonucleotide) which, at a given position, are identical to (i.e. in their ability to form Watson Crick base pairs with the complementary nucleoside) a contiguous nucleotide sequence, at a given position of a separate nucleic acid molecule (e.g. the target nucleic acid). The percentage is calculated by counting the number of aligned bases that are identical between the two sequences, including gaps, dividing by the total number of nucleotides in the oligonucleotide and multiplying by 100.

  • Percent Identity=(Matches×100)/Length of aligned region (with gaps).
  • When determining the identity of the contiguous nucleotide region of an oligonucleotide, the identity is calculated across the length of the contiguous nucleotide region. In embodiments where the entire contiguous nucleotide sequence of the oligonucleotide is the contiguous nucleotide region, identity is therefore calculated across the length of the nucleotide sequence of the oligonucleotide. In this respect the contiguous nucleotide region may be identical to a region of the reference nucleic acid sequence, or in some embodiments may be identical to the entire reference nucleic acid. Unless otherwise indicated a sequence which has 100% identity to a reference sequence is referred to as being identical. For example, the reference sequence may be selected from the group consisting of any one of SEQ ID NOs 5-146 and 156. However, if the oligonucleotide comprises additional nucleotide(s) flanking the contiguous nucleotide region, for example region D′ or D″, these additional flanking nucleotides may be disregarded when determining identity. In some embodiments, identity may be calculated across the entire oligonucleotide sequence.
  • In some embodiments, the antisense oligonucleotide of the invention comprises a contiguous nucleotide region of 10-22 contiguous nucleotides which are identical to SEQ ID NO 156:
  • SEQ ID NO 156: 5′ CAAATATTTACCTGGTTGTTGG 3′
  • In some embodiments, the contiguous nucleotide region consists or comprises of at least 10 contiguous nucleotides, such as 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, contiguous nucleotides, such as from 12-22, such as from 14-18 contiguous nucleotides of SEQ ID NO 156. In some embodiments, the entire contiguous sequence of the oligonucleotide consists or comprises of at least 10 contiguous nucleotides, such as 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, contiguous nucleotides, such as from 12-22, such as from 14-18 contiguous nucleotides of SEQ ID NO 156.
  • In some embodiments, the contiguous nucleotide region is at least 12 contiguous nucleotides of SEQ ID NO 156. In some embodiments, the contiguous nucleotide region is at least 14 contiguous nucleotides of SEQ ID NO 156. In some embodiments, the contiguous nucleotide region is at least 16 contiguous nucleotides SEQ ID NO 156.
  • In some embodiments, the contiguous nucleotide region is at least 10, 12, 14 or 16 contiguous nucleotides which are identical to SEQ ID NO 143.
  • In some embodiments, the contiguous nucleotide region is at least 10, 12, 14 or 16 contiguous nucleotides which are identical to SEQ ID NO 145.
  • In some embodiments, the contiguous nucleotide region is at least 10, 11, 12, 13, 14, 15 or 16 contiguous nucleotides which are identical to SEQ ID NO 143.
  • In some embodiments, the contiguous nucleotide region is at least 10, 11, 12, 13, 14, 15, 16 or 17 contiguous nucleotides which are identical to SEQ ID NO 145.
  • In some embodiments, the contiguous nucleotide consists or comprises SEQ ID NO 143.
  • In some embodiments, the contiguous nucleotide region consists or comprises SEQ ID NO 145.
  • In some embodiments, the contiguous nucleotide region is at least 10, 12, 14 or 16 contiguous nucleotides which are identical to a sequence selected from the group consisting of SEQ ID NO 138, 139, 140, 141, 142, 143, 144 and 145. In some embodiments, the contiguous nucleotide region comprises or consists of a sequence selected from the group consisting of SEQ ID NO 138, 139, 140, 141, 142, 143, 144 and 145.
  • In some embodiments the contiguous nucleotide region comprises the sequence SEQ ID NO 146: TTTACCTGGTT.
  • The invention provides for an antisense oligonucleotide 11-30 nucleotides in length, such as 12-20 nucleotides in length, which comprises the sequence SEQ ID NO 146: TTTACCTGGTT.
  • Hybridization
  • The term “hybridizing” or “hybridizes” as used herein is to be understood as two nucleic acid strands (e.g. an oligonucleotide and a target nucleic acid) forming hydrogen bonds between base pairs on opposite strands thereby forming a duplex. The affinity of the binding between two nucleic acid strands is the strength of the hybridization. It is often described in terms of the melting temperature (Tm) defined as the temperature at which half of the oligonucleotides are duplexed with the target nucleic acid. At physiological conditions Tm is not strictly proportional to the affinity (Mergny and Lacroix, 2003, Oligonucleotides 13:515-537). The standard state Gibbs free energy ΔG° is a more accurate representation of binding affinity and is related to the dissociation constant (Kd) of the reaction by ΔG°=−RTIn(Kd), where R is the gas constant and T is the absolute temperature. Therefore, a very low ΔG° of the reaction between an oligonucleotide and the target nucleic acid reflects a strong hybridization between the oligonucleotide and target nucleic acid. ΔG° is the energy associated with a reaction where aqueous concentrations are 1M, the pH is 7, and the temperature is 37° C. The hybridization of oligonucleotides to a target nucleic acid is a spontaneous reaction and for spontaneous reactions ΔG° is less than zero. ΔG° can be measured experimentally, for example, by use of the isothermal titration calorimetry (ITC) method as described in Hansen et al., 1965, Chem. Comm. 36-38 and Holdgate et al., 2005, Drug Discov Today. The skilled person will know that commercial equipment is available for ΔG° measurements. ΔG° can also be estimated numerically by using the nearest neighbor model as described by SantaLucia, 1998, Proc Natl Mad Sci USA. 95: 1460-1465 using appropriately derived thermodynamic parameters described by Sugimoto et al., 1995, Biochemistry 34:11211-11216 and McTigue et al., 2004, Biochemistry 43:5388-5405. In order to have the possibility of modulating its intended nucleic acid target by hybridization, oligonucleotides of the present invention hybridize to a target nucleic acid with estimated ΔG° values below −10 kcal for oligonucleotides that are 10-30 nucleotides in length. In some embodiments the degree or strength of hybridization is measured by the standard state Gibbs free energy ΔG°. The oligonucleotides may hybridize to a target nucleic acid with estimated ΔG° values below the range of −10 kcal, such as below −15 kcal, such as below −20 kcal and such as below −25 kcal for oligonucleotides that are 8-30 nucleotides in length. In some embodiments the oligonucleotides hybridize to a target nucleic acid with an estimated ΔG° value of −10 to −60 kcal, such as −12 to −40, such as from −15 to −30 kcal or −16 to −27 kcal such as −18 to −25 kcal.
  • Target Sequence
  • The oligonucleotide comprises a contiguous nucleotide region which is complementary to or hybridizes to a sub-sequence of the target nucleic acid molecule. The term “target sequence” as used herein refers to a sequence of nucleotides present in the target nucleic acid which comprises the nucleobase sequence which is complementary to the contiguous nucleotide region or sequence of the oligonucleotide of the invention. In some embodiments, the target sequence consists of a region on the target nucleic acid which is complementary to the contiguous nucleotide region or sequence of the oligonucleotide of the invention. In some embodiments the target sequence is longer than the complementary sequence of a single oligonucleotide, and may, for example represent a preferred region of the target nucleic acid which may be targeted by several oligonucleotides of the invention.
  • The oligonucleotide of the invention comprises a contiguous nucleotide region which is complementary to the target nucleic acid, such as a target sequence.
  • The oligonucleotide comprises a contiguous nucleotide region of at least 10 nucleotides which is complementary to or hybridizes to a target sequence present in the target nucleic acid molecule. The contiguous nucleotide region (and therefore the target sequence) comprises of at least 10 contiguous nucleotides, such as 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, contiguous nucleotides, such as from 12-22, such as from 14-18 contiguous nucleotides.
  • In some embodiments the target sequence is, or is present within SEQ ID NO 147.
  • In some embodiments the target sequence is selected from the group consisting of SEQ ID NO 148, 149, 150, 151, 152, 153, 154 and 155:
  • SEQ ID NO 148: AACAACCAGGTAAATA
    SEQ ID NO 149: CAACCAGGTAAATATTTG
    SEQ ID NO 150: CCAACAACCAGGTAAA
    SEQ ID NO 151: AACCAGGTAAATATTTGG
    SEQ ID NO 152: ACAACCAGGTAAATATTTGG
    SEQ ID NO 153: CAACAACCAGGTAAATAT
    SEQ ID NO 154: ACAACCAGGTAAATAT
    SEQ ID NO 155: AACAACCAGGTAAATAT
  • The invention provides for an antisense oligonucleotide of 10-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of at least 10, contiguous nucleotides which are complementary to a sequence present in a sequence selected from SEQ ID NO 147 & 148-155.
  • The invention provides for an antisense oligonucleotide of 12-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of at least 12 contiguous nucleotides which are complementary to a sequence present in a sequence selected from SEQ ID NO 147 & 148-155.
  • The invention provides for an antisense oligonucleotide of 14-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of at least 14 contiguous nucleotides which are complementary to a sequence present in a sequence selected from SEQ ID NO 147 & 148-155.
  • The invention provides for an antisense oligonucleotide which consists or comprises a contiguous nucleotide region which is complementary to a sequence selected from SEQ ID NO 148-155.
  • The target sequence may be a sub-sequence of the target nucleic acid. In some embodiments the oligonucleotide or contiguous nucleotide region is fully complementary to, or only comprises one or two mismatches to an HTRA1 sub-sequence, such as a sequence selected from the group consisting of SEQ ID NO 148-154. In some embodiments the oligonucleotide or contiguous nucleotide region is fully complementary to, or only comprises one or two mismatches to an HTRA1 sub-sequence SEQ ID NO 147.
  • Target Cell
  • The term a target cell as used herein refers to a cell which is expressing the target nucleic acid. In some embodiments the target cell may be in vivo or in vitro. In some embodiments the target cell is a mammalian cell such as a rodent cell, such as a mouse cell, or a rat cell, or a primate cell such as a monkey cell or a human cell. In some embodiments, the cell may be a pig cell, a dog cell or a rabbit cell. In some embodiments the target cell may be a retinal cell, such as a retinal pigment epithelium (PRE) cell. In some embodiments the cell is selected from the group consisting of RPE cells, Bipolar Cell, Amacrine cells, Endothelial cells, Ganglion cells and Microglia cells. For in vitro assessment, the target cell may be a primary cell or an established cell line, such as U251, ARPE19, HEK293, or rat C6 cells.
  • Target Nucleic Acid
  • According to the present invention, the target nucleic acid is a nucleic acid which encodes mammalian HTRA1 and may for example be a gene, a RNA, a mRNA, and pre-mRNA, a mature mRNA or a cDNA sequence. The target may therefore be referred to as an HTRA1 target nucleic acid.
  • Suitably, the target nucleic acid encodes an HTRA1 protein, in particular mammalian HTRA1, such as human HTRA1 (See for example tables 1 & 2 which provides the mRNA and pre-mRNA sequences for human and rat HTRA1).
  • In some embodiments, the target nucleic acid is selected from the group consisting of SEQ ID NO: 1, 2, 3, and 4, or naturally occurring variants thereof (e.g. sequences encoding a mammalian HTRA1 protein.
  • A target cell is a cell which is expressing the HTRA1 target nucleic acid. In preferred embodiments the target nucleic acid is the HTRA1 mRNA, such as the HTRA1 pre-mRNA or HTRA1 mature mRNA. The poly A tail of HTRA1 mRNA is typically disregarded for antisense oligonucleotide targeting.
  • If employing the oligonucleotide of the invention in research or diagnostics the target nucleic acid may be a cDNA or a synthetic nucleic acid derived from DNA or RNA.
  • The target sequence may be a sub-sequence of the target nucleic acid. In some embodiments the oligonucleotide or contiguous nucleotide region is fully complementary to, or only comprises one or two mismatches to an HTRA1 sub-sequence, such as a sequence selected from the group consisting of SEQ ID NO 148, 149, 150, 151, 152, 153, 154 and 155.
  • Complementarity to the target or sub-sequence thereof is measured over the length of the oligonucleotide, or contiguous nucleotide region thereof.
  • For in vivo or in vitro application, the oligonucleotide of the invention is typically capable of inhibiting the expression of the HTRA1 target nucleic acid in a cell which is expressing the HTRA1 target nucleic acid. The contiguous sequence of nucleobases of the oligonucleotide of the invention is typically complementary to the HTRA1 target nucleic acid, as measured across the length of the oligonucleotide, optionally with the exception of one or two mismatches, and optionally excluding nucleotide based linker regions which may link the oligonucleotide to an optional functional group such as a conjugate, or other non-complementary terminal nucleotides (e.g. region D). The target nucleic acid may, in some embodiments, be a RNA or DNA, such as a messenger RNA, such as a mature mRNA or a pre-mRNA. In some embodiments the target nucleic acid is a RNA or DNA which encodes mammalian HTRA1 protein, such as human HTRA1, e.g. the human HTRA1 mRNA sequence, such as that disclosed as SEQ ID NO 1 (NM_002775.4, GI:190014575). Further information on exemplary target nucleic acids is provided in tables 1 & 2.
  • TABLE 1
    Genome and assembly information for human and rat HTRA1.
    NCBI reference
    Genomic coordinates sequence* accession
    Species Chr. Strand Start End Assembly number for mRNA
    Human
    10 fwd 122461525 122514908 GRCh38.p2 release NM_002775.4
    107
    Rat 1 fwd 201499067 201548508 Rnor_6.0 release NM_031721.1
    105
    Fwd = forward strand.
    The genome coordinates provide the pre-mRNA sequence (genomic sequence).
    The NCBI reference provides the mRNA sequence (cDNA sequence).
    *The National Center for Biotechnology Information reference sequence database is a comprehensive, integrated, non-redundant, well-annotated set of reference sequences including genomic, transcript, and protein. It is hosted at www.ncbi.nlm.nih.gov/refseq.
  • TABLE 2
    Sequence details for human and rat HTRA1.
    Species RNA type Length (nt) SEQ ID NO
    Human mRNA 2138 1
    Human premRNA 53384 2
    Rat mRNA 2012 3
    Rat premRNA 49442 4
  • Naturally Occurring Variant
  • The term “naturally occurring variant” refers to variants of HTRA1 gene or transcripts which originate from the same genetic loci as the target nucleic acid, but may differ for example, by virtue of degeneracy of the genetic code causing a multiplicity of codons encoding the same amino acid, or due to alternative splicing of pre-mRNA, or the presence of polymorphisms, such as single nucleotide polymorphisms, and allelic variants. Based on the presence of the sufficient complementary sequence to the oligonucleotide, the oligonucleotide of the invention may therefore target the target nucleic acid and naturally occurring variants thereof. In some embodiments, the naturally occurring variants have at least 95% such as at least 98% or at least 99% homology to a mammalian HTRA1 target nucleic acid, such as a target nucleic acid selected form the group consisting of SEQ ID NO 1, 2, 3, or 4.
  • Modulation of Expression
  • The term “modulation of expression” as used herein is to be understood as an overall term for an oligonucleotide's ability to alter the amount of HTRA1 when compared to the amount of HTRA1 before administration of the oligonucleotide. Alternatively modulation of expression may be determined by reference to a control experiment where the oligonucleotide of the invention is not administered. One type of modulation is an oligonucleotide's ability to inhibit, down-regulate, reduce, suppress, remove, stop, block, prevent, lessen, lower, avoid or terminate expression of HTRA1, e.g. by degradation of mRNA or blockage of transcription. The antisense oligonucleotide of the invention are capable of inhibiting, down-regulating, reduce, suppress, remove, stop, block, prevent, lessen, lower, avoid or terminate expression of HTRA1.
  • High Affinity Modified Nucleosides
  • A high affinity modified nucleoside is a modified nucleotide which, when incorporated into the oligonucleotide enhances the affinity of the oligonucleotide for its complementary target, for example as measured by the melting temperature (Tm). A high affinity modified nucleoside of the present invention preferably result in an increase in melting temperature between +0.5 to +12° C., more preferably between +1.5 to +10° C. and most preferably between +3 to +8° C. per modified nucleoside. Numerous high affinity modified nucleosides are known in the art and include for example, many 2′ substituted nucleosides as well as locked nucleic acids (LNA) (see e.g. Freier & Altmann; Nucl. Acid Res., 1997, 25, 4429-4443 and Uhlmann; Curr. Opinion in Drug Development, 2000, 3(2), 293-213).
  • Sugar Modifications
  • The oligomer of the invention may comprise one or more nucleosides which have a modified sugar moiety, i.e. a modification of the sugar moiety when compared to the ribose sugar moiety found in DNA and RNA.
  • Numerous nucleosides with modification of the ribose sugar moiety have been made, primarily with the aim of improving certain properties of oligonucleotides, such as affinity and/or nuclease resistance.
  • Such modifications include those where the ribose ring structure is modified, e.g. by replacement with a hexose ring (HNA), or a bicyclic ring, which typically have a biradicle bridge between the C2 and C4 carbons on the ribose ring (LNA), or an unlinked ribose ring which typically lacks a bond between the C2 and C3 carbons (e.g. UNA). Other sugar modified nucleosides include, for example, bicyclohexose nucleic acids (WO2011/017521) or tricyclic nucleic acids (WO2013/154798). Modified nucleosides also include nucleosides where the sugar moiety is replaced with a non-sugar moiety, for example in the case of peptide nucleic acids (PNA), or morpholino nucleic acids.
  • Sugar modifications also include modifications made via altering the substituent groups on the ribose ring to groups other than hydrogen, or the 2′—OH group naturally found in DNA and RNA nucleosides. Substituents may, for example be introduced at the 2′, 3′, 4′ or 5′ positions. Nucleosides with modified sugar moieties also include 2′ modified nucleosides, such as 2′ substituted nucleosides. Indeed, much focus has been spent on developing 2′ substituted nucleosides, and numerous 2′ substituted nucleosides have been found to have beneficial properties when incorporated into oligonucleotides, such as enhanced nucleoside resistance and enhanced affinity.
  • 2′ Modified Nucleosides.
  • A 2′ sugar modified nucleoside is a nucleoside which has a substituent other than H or —OH at the 2′ position (2′ substituted nucleoside) or comprises a 2′ linked biradicle, and includes 2′ substituted nucleosides and LNA (2′-4′ biradicle bridged) nucleosides. For example, the 2′ modified sugar may provide enhanced binding affinity and/or increased nuclease resistance to the oligonucleotide. Examples of 2′ substituted modified nucleosides are 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA (MOE), 2′-amino-DNA, 2′-Fluoro-RNA, and 2′-F-ANA nucleoside. For further examples, please see e.g. Freier & Altmann; Nucl. Acid Res., 1997, 25, 4429-4443 and Uhlmann; Curr. Opinion in Drug Development, 2000, 3(2), 293-213, and Deleavey and Damha, Chemistry and Biology 2012, 19, 937. Below are illustrations of some 2′ substituted modified nucleosides.
  • Figure US20200399641A1-20201224-C00001
  • Locked Nucleic Acid Nucleosides (LNA).
  • LNA nucleosides are modified nucleosides which comprise a linker group (referred to as a biradicle or a bridge) between C2′ and C4′ of the ribose sugar ring of a nucleotide. These nucleosides are also termed bridged nucleic acid or bicyclic nucleic acid (BNA) in the literature.
  • In some embodiments, the modified nucleoside or the LNA nucleosides of the oligomer of the invention has a general structure of the formula I or II:
  • Figure US20200399641A1-20201224-C00002
  • wherein W is selected from —O—, —S—, —N(Ra)—, —C(RaRb)—, such as, in some embodiments —O—;
  • B designates a nucleobase moiety;
  • Z designates an internucleoside linkage to an adjacent nucleoside, or a 5′-terminal group;
  • Z* designates an internucleoside linkage to an adjacent nucleoside, or a 3′-terminal group;
  • X designates a group selected from the list consisting of —C(RaRb)—, —C(Ra)═C(Rb)—, —C(Ra)═N—, —O—, —Si(Ra)2—, —S—, —SO2—, —N(Ra)—, and >C═Z
  • In some embodiments, X is selected from the group consisting of: —O—, —S—, NH—, NRaRb, —CH2—, CRaRb, —C(═CH2)—, and —C(═CRaRb)—
      • In some embodiments, X is —O—
  • Y designates a group selected from the group consisting of —C(RaRb)—, —C(Ra)═C(Rb)—, —C(Ra)═N—, —O—, —Si(Ra)2—, —S—, —SO2—, —N(Ra)—, and >C═Z
  • In some embodiments, Y is selected from the group consisting of: —CH2—, —C(RaRb)—, —CH2CH2—, —C(RaRb)—C(RaRb)—, —CH2CH2CH2—, —C(RaRb)C(RaRb)C(RaRb)—, —C(Ra)═C(Rb)—, and —C(Ra)═N—
  • In some embodiments, Y is selected from the group consisting of: —CH2—, —CHRa—, —CHCH3—, CRaRb
  • or —X—Y— together designate a bivalent linker group (also referred to as a radicle) together designate a bivalent linker group consisting of 1, 2, or 3 groups/atoms selected from the group consisting of —C(RaRb)—, —C(Ra)═C(Rb)—, —C(Ra)═N—, —O—, —Si(Ra)2—, —S—, —SO2—, —N(Ra)—, and >C═Z,
  • In some embodiments, —X—Y— designates a biradicle selected from the groups consisting of: —X—CH2—, —X—CRaRb—, —X—CHRa−, —X—C(HCH3), —O—Y—, —O—CH2—, —S—CH2—, —NH—CH2—, —O—CHCH3—, —CH2—O—CH2, —O—CH(CH3CH3)—, —O—CH2—CH2—, OCH2—CH2—CH2—, —O—CH2OCH2—, —O—NCH2—, —C(═CH2)—CH2—, —NRa—CH2—, N—O—CH2, —S—CRaRb— and —S—CHRa—.
      • In some embodiments —X—Y— designates —O—CH2— or —O—CH(CH3)—.
  • wherein Z is selected from —O—, —S—, and —N(Ra)—,
  • and Ra and, when present Rb, each is independently selected from hydrogen, optionally substituted C1-6-alkyl, optionally substituted C2-6-alkenyl, optionally substituted C2-6-alkynyl, hydroxy, optionally substituted C1-6-alkoxy, C2-6-alkoxyalkyl, C2-6-alkenyloxy, carboxy, C1-6-alkoxycarbonyl, C1-6-alkylcarbonyl, formyl, aryl, aryloxy-carbonyl, aryloxy, arylcarbonyl, heteroaryl, heteroaryloxy-carbonyl, heteroaryloxy, heteroarylcarbonyl, amino, mono- and di(C1-6-alkyl)amino, carbamoyl, mono- and di(C1-6-alkyl)-amino-carbonyl, amino-C1-6-alkyl-aminocarbonyl, mono- and di(C1-6-alkyl)amino-C1-6-alkyl-aminocarbonyl, C1-6-alkyl-carbonylamino, carbamido, C1-6-alkanoyloxy, sulphono, C1-6-alkylsulphonyloxy, nitro, azido, sulphanyl, C1-6-alkylthio, halogen, where aryl and heteroaryl may be optionally substituted and where two geminal substituents Ra and Rb together may designate optionally substituted methylene (═CH2), wherein for all chiral centers, asymmetric groups may be found in either R or S orientation.
  • wherein R1, R2, R3, R5 and R5* are independently selected from the group consisting of: hydrogen, optionally substituted C1-6-alkyl, optionally substituted C2-6-alkenyl, optionally substituted C2-6-alkynyl, hydroxy, C1-6-alkoxy, C2-6-alkoxyalkyl, C2-6-alkenyloxy, carboxy, C1-6-alkoxycarbonyl, C1-6-alkylcarbonyl, formyl, aryl, aryloxy-carbonyl, aryloxy, arylcarbonyl, heteroaryl, heteroaryloxy-carbonyl, heteroaryloxy, heteroarylcarbonyl, amino, mono- and di(C1-6-alkyl)amino, carbamoyl, mono- and di(C1-6-alkyl)-amino-carbonyl, amino-C1-6-alkyl-aminocarbonyl, mono- and di(C1-6-alkyl)amino-C1-6-alkyl-aminocarbonyl, C1-6-alkyl-carbonylamino, carbamido, C1-6-alkanoyloxy, sulphono, C1-6-alkylsulphonyloxy, nitro, azido, sulphanyl, C1-6-alkylthio, halogen, where aryl and heteroaryl may be optionally substituted, and where two geminal substituents together may designate oxo, thioxo, imino, or optionally substituted methylene.
  • In some embodiments R1, R2, R3, R5 and R5* are independently selected from C1-6 alkyl, such as methyl, and hydrogen.
      • In some embodiments R1, R2, R3, R5 and R5* are all hydrogen.
  • In some embodiments R1, R2, R3, are all hydrogen, and either R5 and R5* is also hydrogen and the other of R5 and R5* is other than hydrogen, such as C1-6 alkyl such as methyl.
  • In some embodiments, Ra is either hydrogen or methyl. In some embodiments, when present, Rb is either hydrogen or methyl.
      • In some embodiments, one or both of Ra and Rb is hydrogen
  • In some embodiments, one of Ra and Rb is hydrogen and the other is other than hydrogen
      • In some embodiments, one of Ra and Rb is methyl and the other is hydrogen
      • In some embodiments, both of Ra and Rb are methyl.
  • In some embodiments, the biradicle —X—Y— is —O—CH2—, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. Such LNA nucleosides are disclosed in WO99/014226, WO00/66604, WO98/039352 and WO2004/046160 which are all hereby incorporated by reference, and include what are commonly known as beta-D-oxy LNA and alpha-L-oxy LNA nucleosides.
  • In some embodiments, the biradicle —X—Y— is —S—CH2—, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. Such thio LNA nucleosides are disclosed in WO99/014226 and WO2004/046160 which are hereby incorporated by reference.
  • In some embodiments, the biradicle —X—Y— is —NH—CH2—, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. Such amino LNA nucleosides are disclosed in WO99/014226 and WO2004/046160 which are hereby incorporated by reference.
  • In some embodiments, the biradicle —X—Y— is —O—CH2—CH2— or —O—CH2—CH2—CH2—, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. Such LNA nucleosides are disclosed in WO00/047599 and Morita et al, Bioorganic & Med. Chem. Lett. 12 73-76, which are hereby incorporated by reference, and include what are commonly known as 2′-O-4′C-ethylene bridged nucleic acids (ENA).
  • In some embodiments, the biradicle —X—Y— is —O—CH2—, W is O, and all of R1, R2, R3, and one of R5 and R5* are hydrogen, and the other of R5 and R5* is other than hydrogen such as C1-6 alkyl, such as methyl. Such 5′ substituted LNA nucleosides are disclosed in WO2007/134181 which is hereby incorporated by reference.
  • In some embodiments, the biradicle —X—Y— is —O—CRaRb—, wherein one or both of Ra and Rb are other than hydrogen, such as methyl, W is O, and all of R1, R2, R3, and one of R5 and R5* are hydrogen, and the other of R5 and R5* is other than hydrogen such as C1-6 alkyl, such as methyl. Such bis modified LNA nucleosides are disclosed in WO2010/077578 which is hereby incorporated by reference.
  • In some embodiments, the biradicle —X—Y— designate the bivalent linker group —O—CH(CH2OCH3)— (2′ O-methoxyethyl bicyclic nucleic acid—Seth at al., 2010, J. Org. Chem. Vol 75(5) pp. 1569-81). In some embodiments, the biradicle —X—Y— designate the bivalent linker group —O—CH(CH2CH3)— (2′O-ethyl bicyclic nucleic acid—Seth at al., 2010, J. Org. Chem. Vol 75(5) pp. 1569-81). In some embodiments, the biradicle —X—Y— is —O—CHRa—, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. Such 6′ substituted LNA nucleosides are disclosed in WO10036698 and WO07090071 which are both hereby incorporated by reference.
  • In some embodiments, the biradicle —X—Y— is —O—CH(CH2OCH3)—, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. Such LNA nucleosides are also known as cyclic MOEs in the art (cMOE) and are disclosed in WO07090071.
  • In some embodiments, the biradicle —X—Y— designate the bivalent linker group —O—CH(CH3)—. — in either the R— or S— configuration. In some embodiments, the biradicle —X—Y— together designate the bivalent linker group —O—CH2—O—CH2— (Seth at al., 2010, J. Org. Chem). In some embodiments, the biradicle —X—Y— is —O—CH(CH3)—, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. Such 6′ methyl LNA nucleosides are also known as cET nucleosides in the art, and may be either (S)cET or (R)cET stereoisomers, as disclosed in WO07090071 (beta-D) and WO2010/036698 (alpha-L) which are both hereby incorporated by reference).
  • In some embodiments, the biradicle —X—Y— is —O—CRaRb—, wherein in neither Ra or Rb is hydrogen, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. In some embodiments, Ra and Rb are both methyl. Such 6′ di-substituted LNA nucleosides are disclosed in WO 2009006478 which is hereby incorporated by reference.
  • In some embodiments, the biradicle —X—Y— is —S—CHRa—, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. Such 6′ substituted thio LNA nucleosides are disclosed in WO11156202 which is hereby incorporated by reference. In some 6′ substituted thio LNA embodiments Ra is methyl.
  • In some embodiments, the biradicle —X—Y— is —C(═CH2)—C(RaRb)—, such as —C(═CH2)—CH2—, or —C(═CH2)—CH(CH3)—W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. Such vinyl carbo LNA nucleosides are disclosed in WO08154401 and WO09067647 which are both hereby incorporated by reference.
  • In some embodiments the biradicle —X—Y— is —N(—ORa)—, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. In some embodiments Ra is C1-6 alkyl such as methyl. Such LNA nucleosides are also known as N substituted LNAs and are disclosed in WO2008/150729 which is hereby incorporated by reference. In some embodiments, the biradicle —X—Y— together designate the bivalent linker group —O—NRa—CH3— (Seth at al., 2010, J. Org. Chem). In some embodiments the biradicle —X—Y— is —N(Ra)—, W is O, and all of R1, R2, R3, R5 and R5* are all hydrogen. In some embodiments Ra is C1-6 alkyl such as methyl.
  • In some embodiments, one or both of R5 and R5* is hydrogen and, when substituted the other of R5 and R5* is C1-6 alkyl such as methyl. In such an embodiment, R1, R2, R3, may all be hydrogen, and the biradicle —X—Y— may be selected from —O—CH2— or —O—C(HCRa)—, such as —O—C(HCH3)—.
  • In some embodiments, the biradicle is —CRaRb—O—CRaRb—, such as CH2—O—CH2—, W is O and all of R1, R2, R3, R5 and R5* are all hydrogen. In some embodiments Ra is C1-6 alkyl such as methyl. Such LNA nucleosides are also known as conformationally restricted nucleotides (CRNs) and are disclosed in WO2013036868 which is hereby incorporated by reference.
  • In some embodiments, the biradicle is —O—CRaRb—O—CRaRb—, such as O—CH2—O—CH2—, W is O and all of R1, R2, R3, R5 and R5* are all hydrogen. In some embodiments Ra is C1-6 alkyl such as methyl. Such LNA nucleosides are also known as COC nucleotides and are disclosed in Mitsuoka et al., Nucleic Acids Research 2009 37(4), 1225-1238, which is hereby incorporated by reference.
  • It will be recognized than, unless specified, the LNA nucleosides may be in the beta-D or alpha-L stereoisoform.
  • Examples of LNA nucleosides are presented in Scheme 1.
  • Figure US20200399641A1-20201224-C00003
  • As illustrated in the examples, in some embodiments of the invention the LNA nucleosides in the oligonucleotides are beta-D-oxy-LNA nucleosides.
  • Nuclease Mediated Degradation
  • Nuclease mediated degradation refers to an oligonucleotide capable of mediating degradation of a complementary nucleotide sequence when forming a duplex with such a sequence.
  • In some embodiments, the oligonucleotide may function via nuclease mediated degradation of the target nucleic acid, where the oligonucleotides of the invention are capable of recruiting a nuclease, particularly and endonuclease, preferably endoribonuclease (RNase), such as RNase H. Examples of oligonucleotide designs which operate via nuclease mediated mechanisms are oligonucleotides which typically comprise a region of at least 5 or 6 DNA nucleosides and are flanked on one side or both sides by affinity enhancing nucleosides, for example gapmers, headmers and tailmers.
  • RNase H Activity and Recruitment
  • The RNase H activity of an antisense oligonucleotide refers to its ability to recruit RNase H when in a duplex with a complementary RNA molecule. WO01/23613 provides in vitro methods for determining RNaseH activity, which may be used to determine the ability to recruit RNaseH. Typically an oligonucleotide is deemed capable of recruiting RNase H if it, when provided with a complementary target nucleic acid sequence, has an initial rate, as measured in pmol/l/min, of at least 5%, such as at least 10% or more than 20% of the of the initial rate determined when using a oligonucleotide having the same base sequence as the modified oligonucleotide being tested, but containing only DNA monomers, with phosphorothioate linkages between all monomers in the oligonucleotide, and using the methodology provided by Example 91-95 of WO01/23613 (hereby incorporated by reference).
  • Gapmer
  • The term gapmer as used herein refers to an antisense oligonucleotide which comprises a region of RNase H recruiting oligonucleotides (gap) which is flanked 5′ and 3′ by regions which comprise one or more affinity enhancing modified nucleosides (flanks or wings). Various gapmer designs are described herein. Headmers and tailmers are oligonucleotides capable of recruiting RNase H where one of the flanks is missing, i.e. only one of the ends of the oligonucleotide comprises affinity enhancing modified nucleosides. For headmers the 3′ flank is missing (i.e. the 5′ flank comprises affinity enhancing modified nucleosides) and for tailmers the 5′ flank is missing (i.e. the 3′ flank comprises affinity enhancing modified nucleosides).
  • LNA Gapmer
  • The term LNA gapmer is a gapmer oligonucleotide wherein at least one of the affinity enhancing modified nucleosides is an LNA nucleoside.
  • Mixed Wing Gapmer
  • The term mixed wing gapmer refers to a LNA gapmer wherein the flank regions comprise at least one LNA nucleoside and at least one non-LNA modified nucleoside, such as at least one DNA nucleoside or at least one 2′ substituted modified nucleoside, such as, for example, 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA (MOE), 2′-amino-DNA, 2′-Fluoro-RNA and 2′-F-ANA nucleoside(s). In some embodiments the mixed wing gapmer has one flank which comprises LNA nucleosides (e.g. 5′ or 3′) and the other flank (3′ or 5′ respectfully) comprises 2′ substituted modified nucleoside(s).
  • Conjugate
  • The term conjugate as used herein refers to an oligonucleotide which is covalently linked to a non-nucleotide moiety (conjugate moiety or region C or third region).
  • The term conjugate as used herein refers to an oligonucleotide which is covalently linked to a non-nucleotide moiety (conjugate moiety or region C or third region).
  • In some embodiments, the non-nucleotide moiety selected from the group consisting of a protein, such as an enzyme, an antibody or an antibody fragment or a peptide; a lipophilic moiety such as a lipid, a phospholipid, a sterol; a polymer, such as polyethyleneglycol or polypropylene glycol; a receptor ligand; a small molecule; a reporter molecule; and a non-nucleosidic carbohydrate.
  • Linkers
  • A linkage or linker is a connection between two atoms that links one chemical group or segment of interest to another chemical group or segment of interest via one or more covalent bonds. Conjugate moieties can be attached to the oligonucleotide directly or through a linking moiety (e.g. linker or tether). Linkers serve to covalently connect a third region, e.g. a conjugate moiety to an oligonucleotide (e.g. the termini of region A or C).
  • In some embodiments of the invention the conjugate or oligonucleotide conjugate of the invention may optionally, comprise a linker region which is positioned between the oligonucleotide and the conjugate moiety. In some embodiments, the linker between the conjugate and oligonucleotide is biocleavable.
  • Biocleavable linkers comprising or consisting of a physiologically labile bond that is cleavable under conditions normally encountered or analogous to those encountered within a mammalian body. Conditions under which physiologically labile linkers undergo chemical transformation (e.g., cleavage) include chemical conditions such as pH, temperature, oxidative or reductive conditions or agents, and salt concentration found in or analogous to those encountered in mammalian cells. Mammalian intracellular conditions also include the presence of enzymatic activity normally present in a mammalian cell such as from proteolytic enzymes or hydrolytic enzymes or nucleases. In one embodiment the biocleavable linker is susceptible to S1 nuclease cleavage. In a preferred embodiment the nuclease susceptible linker comprises between 1 and 10 nucleosides, such as 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 nucleosides, more preferably between 2 and 6 nucleosides and most preferably between 2 and 4 linked nucleosides comprising at least two consecutive phosphodiester linkages, such as at least 3 or 4 or 5 consecutive phosphodiester linkages. Preferably the nucleosides are DNA or RNA. Phosphodiester containing biocleavable linkers are described in more detail in WO 2014/076195 (hereby incorporated by reference), and may be referred to as region D herein.
  • Conjugates may also be linked to the oligonucleotide via non biocleavable linkers, or in some embodiments the conjugate may comprise a non-cleavable linker which is covalently attached to the biocleavable linker. Linkers that are not necessarily biocleavable but primarily serve to covalently connect a conjugate moiety to an oligonucleotide or biocleavable linker. Such linkers may comprise a chain structure or an oligomer of repeating units such as ethylene glycol, amino acid units or amino alkyl groups. In some embodiments the linker (region Y) is an amino alkyl, such as a C2-C36 amino alkyl group, including, for example C6 to C12 amino alkyl groups. In some embodiments the linker (region Y) is a C6 amino alkyl group. Conjugate linker groups may be routinely attached to an oligonucleotide via use of an amino modified oligonucleotide, and an activated ester group on the conjugate group.
  • Treatment
  • The term ‘treatment’ as used herein refers to both treatment of an existing disease (e.g. a disease or disorder as herein referred to), or prevention of a disease, i.e. prophylaxis. It will therefore be recognized that treatment as referred to herein may, in some embodiments, be prophylactic.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The Oligonucleotides of the Invention
  • The invention relates to oligonucleotides capable of inhibiting the expression of HTRA1. The modulation is may achieved by hybridizing to a target nucleic acid encoding HTRA1 or which is involved in the regulation of HTRA1. The target nucleic acid may be a mammalian HTRA 1 sequence, such as a sequence selected from the group consisting of SEQ ID 1, 2, 3 or 4.
  • The oligonucleotide of the invention is an antisense oligonucleotide which targets HTRA1, such as a mammalian HTRA1.
  • In some embodiments the antisense oligonucleotide of the invention is capable of modulating the expression of the target by inhibiting or down-regulating it. Preferably, such modulation produces an inhibition of expression of at least 20% compared to the normal expression level of the target, such as at least 30%, 40%, 50%, 60%, 70%, 80%, or 90% inhibition compared to the normal expression level of the target. In some embodiments compounds of the invention may be capable of inhibiting expression levels of HTRA1 mRNA by at least 60% or 70% in vitro using ARPE-19 cells. In some embodiments compounds of the invention may be capable of inhibiting expression levels of HTRA1 mRNA by at least 60% or 70% in vitro using ARPE-19 cells. In some embodiments compounds of the invention may be capable of inhibiting expression levels of HTRA1 protein by at least 50% in vitro using ARPE-19 cells. Suitably, the examples provide assays which may be used to measure HTRA1 RNA or protein inhibition (e.g. example 3). The target modulation is triggered by the hybridization between a contiguous nucleotide sequence of the oligonucleotide and the target nucleic acid. In some embodiments the oligonucleotide of the invention comprises mismatches between the oligonucleotide and the target nucleic acid. Despite mismatches hybridization to the target nucleic acid may still be sufficient to show a desired modulation of HTRA1 expression. Reduced binding affinity resulting from mismatches may advantageously be compensated by increased number of nucleotides in the oligonucleotide and/or an increased number of modified nucleosides capable of increasing the binding affinity to the target, such as 2′ modified nucleosides, including LNA, present within the oligonucleotide sequence.
  • An aspect of the present invention relates to an antisense oligonucleotide which comprises a contiguous nucleotide region of 10 to 30 nucleotides in length with at least 90% complementarity to HTRA1 target sequence, such as fully complementary to an HTRA1 target sequence, e.g. a nucleic acid selected from the group consisting SEQ ID NO 1, 2, 3 & 4.
  • In some embodiments, the oligonucleotide comprises a contiguous sequence which is at least 90% complementary, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, or 100% complementary with a region of the target nucleic acid.
  • In some embodiments, the oligonucleotide of the invention, or a contiguous nucleotide sequence thereof is fully complementary (100% complementary) to a region of the target nucleic acid, or in some embodiments may comprise one or two mismatches between the oligonucleotide and the target nucleic acid.
  • In some embodiments the oligonucleotide, or a contiguous nucleotide sequence of at least 12 nucleotides thereof, is at least 90% complementary, such as fully (or 100%) complementary to a region of SEQ ID NO 147.
  • In some embodiments the oligonucleotide, or a contiguous nucleotide sequence of at least 12 nucleotides thereof, is at least 90% complementary, such as fully (or 100%) complementary to a region of a sequence selected from the group consisting of SEQ ID NOs 148, 149, 150, 151, 152, 153, 154 and 155.
  • In some embodiments the oligonucleotide, or a contiguous nucleotide sequence of at least 14 nucleotides thereof, is fully (or 100%) complementary to SEQ ID 147, or a sequence selected from the group consisting of SEQ ID NOs 148, 149, 150, 151, 152, 153, 154 and 155.
  • In some embodiments the oligonucleotide, or a contiguous nucleotide sequence of at least 16 nucleotides thereof, is fully (or 100%) complementary to SEQ ID 147, or a sequence selected from the group consisting of SEQ ID NOs 148, 149, 150, 151, 152, 153, 154 and 155.
  • In some embodiments the oligonucleotide, or contiguous nucleotide region thereof is fully (or 100%) complementary to a sequence selected from the group consisting of SEQ ID NOs 148, 149, 150, 151, 152, 153, 154 and 155.
  • In some embodiments, the oligonucleotide or contiguous nucleotide region thereof comprises or consists of a sequence selected from the group consisting of SEQ ID NOs 143, 138, 139, 140, 141, 142, 144 and 145:
  • SEQ ID NO 143: TATTTACCTGGTTGTT
    SEQ ID NO 138: CAAATATTTACCTGGTTG
    SEQ ID NO 139: TTTACCTGGTTGTTGG
    SEQ ID NO 140: CCAAATATTTACCTGGTT
    SEQ ID NO 141: CCAAATATTTACCTGGTTGT
    SEQ ID NO 142: ATATTTACCTGGTTGTTG
    SEQ ID NO 144: ATATTTACCTGGTTGT
    SEQ ID NO 145: ATATTTACCTGGTTGTT
  • It is understood that the oligonucleotide motif sequences can be modified to for example increase nuclease resistance and/or binding affinity to the target nucleic acid. Modifications are described in the definitions and in the “Oligonucleotide design” section.
  • In some embodiments, the oligonucleotide of the invention, or contiguous nucleotide region thereof is fully complementary (100% complementary) to a region of the target nucleic acid, or in some embodiments may comprise one or two mismatches between the oligonucleotide and the target nucleic acid. In some embodiments the oligonucleotide, or contiguous nucleotide sequence of at least 12 nucleotides thereof, is at least 90% complementary, such as fully (or 100%) complementary to the target nucleic acid sequence.
  • In some embodiments the oligonucleotide, or a contiguous nucleotide sequence of at least 12 nucleotides thereof, has 100% identity to a sequence selected from the group consisting of SEQ ID NOs 5-107, or SEQ ID NOs 108-137.
  • In some embodiments the oligonucleotide, or a contiguous nucleotide sequence of at least 14 nucleotides thereof, has 100% identity to a sequence selected from the group consisting of SEQ ID NOs 5-107, or SEQ ID NOs 108-137.
  • In some embodiments the oligonucleotide, or contiguous nucleotide sequence of at least 16 nucleotides thereof, has 100% identity to a sequence selected from the group consisting of SEQ
  • ID NOs 5-107, or SEQ ID NOs 108-137.
  • In some embodiments the oligonucleotide, or contiguous nucleotide region thereof, comprises or consists of a sequence selected from SEQ ID NOs 5-107, or SEQ ID NOs 108-137.
  • In some embodiments the compound of the invention is selected from the group consisting of:
  • SEQ
    ID Compound
    NO ID #
    138 138.1 mCsAsAsAstsastststsascscstsgsGsTsTsG
    139 139.1 TsTstsascscstsgsgststsgstsTsGsG
    140 140.1 mCs mCsAsAsastsastststsascscstsgsGsTsT
    141 141.1 mCs mCsAsasastsastststsascscstsgsgststsGsT
    142 142.1 AsTsAstststsascscstsgsgststsgsTsTsG
    143 143.1 TsAsTststsascscstsgsgstsTsGsTsT
    143 143.2 TsAstststsascscstsgsGstsTsgsTsT
    143 143.3 TsAsTststsascscstsgsgsTsTsgsTsT
    144 144.1 AsTsAsTststsascscstsgsgsTsTsGsT
    144 144.2 AstsAsTsTstsascscstsgsgstsTsGsT
    145 145.1 AstsAsTststsascscstsgsGsTsTsgsTsT
    145 145.2 AsTsAstststsascscstsgsGstsTsgsTsT
    145 145.3 AstsAsTststsascscstsgsgstsTsGsTsT
  • Wherein capital letters represent beta-D-oxy LNA nucleosides, all LNA cytosines are 5-methyl cytosine (as indicated by the superscript m), lower case letters represent DNA nucleosides. All internucleoside linkages are phosphorothioate internucleoside linkages (as indicated by the subscript s).
  • Oligonucleotide Design
  • Oligonucleotide design refers to the pattern of nucleoside sugar modifications in the oligonucleotide sequence. The oligonucleotides of the invention comprise sugar-modified nucleosides and may also comprise DNA or RNA nucleosides. In some embodiments, the oligonucleotide comprises sugar-modified nucleosides and DNA nucleosides. Incorporation of modified nucleosides into the oligonucleotide of the invention may enhance the affinity of the oligonucleotide for the target nucleic acid. In that case, the modified nucleosides can be referred to as affinity enhancing modified nucleotides.
  • In an embodiment, the oligonucleotide comprises at least 1 modified nucleoside, such as at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15 or at least 16 modified nucleosides. In an embodiment the oligonucleotide comprises from 1 to 10 modified nucleosides, such as from 2 to 9 modified nucleosides, such as from 3 to 8 modified nucleosides, such as from 4 to 7 modified nucleosides, such as 6 or 7 modified nucleosides. In an embodiment, the oligonucleotide of the invention may comprise modifications, which are independently selected from these three types of modifications (modified sugar, modified nucleobase and modified internucleoside linkage) or a combination thereof. Preferably the oligonucleotide comprises one or more sugar modified nucleosides, such as 2′ sugar modified nucleosides. Preferably the oligonucleotide of the invention comprise the one or more 2′ sugar modified nucleoside independently selected from the group consisting of 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA, 2′-amino-DNA, 2′-fluoro-DNA, arabino nucleic acid (ANA), 2′-fluoro-ANA and LNA nucleosides. Even more preferably the one or more modified nucleoside is LNA.
  • In some embodiments, at least 1 of the modified nucleosides is a locked nucleic acid (LNA), such as at least 2, such as at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 of the modified nucleosides are LNA. In a still further embodiment all the modified nucleosides are LNA.
  • In a further embodiment the oligonucleotide comprises at least one modified internucleoside linkage. In a preferred embodiment the internucleoside linkages within the contiguous nucleotide sequence are phosphorothioate or boranophosphate internucleoside linkages. In some embodiments all the internucleotide linkages in the contiguous sequence of the oligonucleotide are phosphorothioate linkages.
  • In some embodiments, the oligonucleotide of the invention comprise at least one modified nucleoside which is a 2′-MOE-RNA, such as 2, 3, 4, 5, 6, 7, 8, 9 or 10 2′-MOE-RNA nucleoside units. In some embodiments, at least one of said modified nucleoside is 2′-fluoro DNA, such as 2, 3, 4, 5, 6, 7, 8, 9 or 10 2′-fluoro-DNA nucleoside units.
  • In some embodiments, the oligonucleotide of the invention comprises at least one LNA unit, such as 1, 2, 3, 4, 5, 6, 7, or 8 LNA units, such as from 2 to 6 LNA units, such as from 3 to 7 LNA units, 4 to 8 LNA units or 3, 4, 5, 6 or 7 LNA units. In some embodiments, all the modified nucleosides are LNA nucleosides. In some embodiments, all LNA cytosine units are 5-methyl-cytosine. In some embodiments the oligonucleotide or contiguous nucleotide region thereof has at least 1 LNA unit at the 5′ end and at least 2 LNA units at the 3′ end of the nucleotide sequence. In some embodiments all cytosine nucleobases present in the oligonucleotide of the invention are 5-methyl-cytosine.
  • In some embodiments, the oligonucleotide of the invention comprises at least one LNA unit and at least one 2′ substituted modified nucleoside.
  • In some embodiments of the invention, the oligonucleotide comprise both 2′ sugar modified nucleosides and DNA units.
  • In an embodiment of the invention the oligonucleotide of the invention is capable of recruiting RNase H.
  • In some embodiments, the oligonucleotide of the invention or contiguous nucleotide region thereof is a gapmers oligonucleotide.
  • Gapmer Design
  • In some embodiments the oligonucleotide of the invention, or contiguous nucleotide region thereof, has a gapmer design or structure also referred herein merely as “Gapmer”. In a gapmer structure the oligonucleotide comprises at least three distinct structural regions a 5′-flank, a gap and a 3′-flank, F-G-F′ in ‘5->3’ orientation. In this design, flanking regions F and F′ (also termed wing regions) comprise at least one sugar modified nucleoside which is adjacent to region G, and may in some embodiments comprise a contiguous stretch of 2-7 sugar modified nucleoside, or a contiguous stretch of sugar modified and DNA nucleosides (mixed wings comprising both sugar modified and DNA nucleosides). Consequently, the nucleosides of the 5′ flanking region and the 3′ flanking region which are adjacent to the gap region are sugar modified nucleosides, such as 2′ modified nucleosides. The gap region, G, comprises a contiguous stretch of nucleotides which are capable of recruiting RNase H, when the oligonucleotide is in duplex with the HTRA1target nucleic acid. In some embodiments, region G comprises a contiguous stretch of 5-16 DNA nucleosides. The gapmer region F-G-F′ is complementary to the HTRA1 target nucleic acid, and may therefore be the contiguous nucleotide region of the oligonucleotide.
  • Regions F and F′, flanking the 5′ and 3′ ends of region G, may comprise one or more affinity enhancing modified nucleosides. In some embodiments, the 3′ flank comprises at least one LNA nucleoside, preferably at least 2 LNA nucleosides. In some embodiments, the 5′ flank comprises at least one LNA nucleoside. In some embodiments both the 5′ and 3′ flanking regions comprise a LNA nucleoside. In some embodiments all the nucleosides in the flanking regions are LNA nucleosides. In other embodiments, the flanking regions may comprise both LNA nucleosides and other nucleosides (mixed flanks), such as DNA nucleosides and/or non-LNA modified nucleosides, such as 2′ substituted nucleosides. In this case the gap is defined as a contiguous sequence of at least 5 RNase H recruiting nucleosides (such as 5-16 DNA nucleosides) flanked at the 5′ and 3′ end by an affinity enhancing modified nucleoside, such as an LNA, such as beta-D-oxy-LNA.
  • Region F
  • Region F (5′ flank or 5′ wing) attached to the ′5 end of region G comprises, contains or consists of at least one sugar modified nucleoside such as at least 2, at least 3, at least 4, at least 5, at least 6, at least 7 modified nucleosides. In some embodiments region F comprises or consists of from 1 to 7 modified nucleosides, such as from 2 to 6 modified nucleosides, such as from 2 to 5 modified nucleosides, such as from 2 to 4 modified nucleosides, such as from 1 to 3 modified nucleosides, such as 1, 2, 3 or 4 modified nucleosides.
  • In an embodiment, one or more or all of the modified nucleosides in region F are 2′ modified nucleosides.
  • In a further embodiment one or more of the 2′ modified nucleosides in region F are selected from 2′-O-alkyl-RNA units, 2′-O-methyl-RNA, 2′-amino-DNA units, 2′-fluoro-DNA units, 2′-alkoxy-RNA, MOE units, LNA units, arabino nucleic acid (ANA) units and 2′-fluoro-ANA units.
  • In one embodiment of the invention all the modified nucleosides in region F are LNA nucleosides. In a further embodiment the LNA nucleosides in region F are independently selected from the group consisting of oxy-LNA, thio-LNA, amino-LNA, cET, and/or ENA, in either the beta-D or alpha-L configurations or combinations thereof. In a preferred embodiment region F has at least 1 beta-D-oxy LNA unit, at the 5′ end of the contiguous sequence.
  • Region G
  • Region G (gap region) may comprise, contain or consist of at 5-16 consecutive DNA nucleosides capable of recruiting RNaseH. In a further embodiment region G comprise, contain or consist of from 5 to 12, or from 6 to 10 or from 7 to 9, such as 8 consecutive nucleotide units capable of recruiting RNaseH.
  • In a still further embodiment at least one nucleoside unit in region G is a DNA nucleoside unit, such as from 4 to 20 or 6 to 18 DNA units, such as 5 to 16, In some embodiments, all of the nucleosides of region G are DNA units.
  • In further embodiments the region G may consist of a mixture of DNA and other nucleosides capable of mediating RNase H cleavage. In some embodiments, at least 50% of the nucleosides of region G are DNA, such as at least 60%, at least 70% or at least 80%, or at least 90% DNA.
  • Region F′
  • Region F′ (3′ flank or 3′ wing) attached to the ′3 end of region G comprises, contains or consists of at least one sugar modified nucleoside such as at least 2, at least 3, at least 4, at least 5, at least 6, at least 7 modified nucleosides. In some embodiments region F′ comprises or consists of from 1 to 7 modified nucleosides, such as from 2 to 6 modified nucleosides, such as from 2 to 5 modified nucleosides, such as from 2 to 4 modified nucleosides, such as from 1 to 3 modified nucleosides, such as 1, 2, 3 or 4 modified nucleosides.
  • In an embodiment, one or more or all of the modified nucleosides in region F′ are 2′ modified nucleosides.
  • In a further embodiment one or more of the 2′ modified nucleosides in region F′ are selected from 2′-O-alkyl-RNA units, 2′-O-methyl-RNA, 2′-amino-DNA units, 2′-fluoro-DNA units, 2′-alkoxy-RNA, MOE units, LNA units, arabino nucleic acid (ANA) units and 2′-fluoro-ANA units.
  • In one embodiment of the invention all the modified nucleosides in region F′ are LNA nucleosides. In a further embodiment the LNA nucleosides in region F′ are independently selected from the group consisting of oxy-LNA, thio-LNA, amino-LNA, cET, and/or ENA, in either the beta-D or alpha-L configurations or combinations thereof. In a preferred embodiment region F′ has at least 1 beta-D-oxy LNA unit, at the 5′ end of the contiguous sequence.
  • Region D, D′ and D″
  • The oligonucleotide of the invention n comprises a contiguous nucleotide region which is complementary to the target nucleic acid. In some embodiments, the oligonucleotide may further comprise additional nucleotides positioned 5′ and/or 3′ to the contiguous nucleotide region, which are referred to as region D herein. Region D′ and D″ can be attached to the 5′ end of region F or the 3′ end of region F′, respectively. The D regions (region D′ or D″) may in some embodiments form part of the contiguous nucleotide sequence which is complementary to the target nucleic acid, or in other embodiments the D region(s) may be non-complementary to the target nucleic acid.
  • In some embodiments the oligonucleotide of the invention consists or comprises of the contiguous nucleotide region and optionally 1-5 additional 5′ nucleotides (region D′).
  • In some embodiments the oligonucleotide of the invention consists or comprises of the contiguous nucleotide region and optionally 1-5 additional 3′ nucleotides (region D″).
  • Region D′ or D″ may independently comprise 1, 2, 3, 4 or 5 additional nucleotides, which may be complementary or non-complementary to the target nucleic acid. In this respect the oligonucleotide of the invention, may in some embodiments comprise a contiguous nucleotide sequence capable of modulating the target which is flanked at the 5′ and/or 3′ end by additional nucleotides. Such additional nucleotides may serve as a nuclease susceptible biocleavable linker, and may therefore be used to attach a functional group such as a conjugate moiety to the oligonucleotide of the invention. In some embodiments the additional 5′ and/or 3′ end nucleotides are linked with phosphodiester linkages, and may be DNA or RNA. In another embodiment, the additional 5′ and/or 3′ end nucleotides are modified nucleotides which may for example be included to enhance nuclease stability or for ease of synthesis. In some embodiments the oligonucleotide of the invention comprises a region D′ and/or D″ in addition to the contiguous nucleotide region.
  • In some embodiments, the gapmer oligonucleotide of the present invention can be represented by the following formulae:

  • F-G-F′; in particular F1-7-G4-12-F′1-7

  • D′-F-G-F′, in particular D′1-3—F1-7-G4-12-F′1-7

  • F-G-F′-D″, in particular F1-7-G4-12-F′1-7-D″1-3

  • D′-F-G-F′-D″, in particular D′1-3—F1-7-G4-12-F′1-7-D″1-3
  • Method of Manufacture
  • In a further aspect, the invention provides methods for manufacturing the oligonucleotides of the invention comprising reacting nucleotide units and thereby forming covalently linked contiguous nucleotide units comprised in the oligonucleotide. Preferably, the method uses phophoramidite chemistry (see for example Caruthers et al, 1987, Methods in Enzymology vol. 154, pages 287-313). In a further embodiment the method further comprises reacting the contiguous nucleotide sequence with a conjugating moiety (ligand). In a further aspect a method is provided for manufacturing the composition of the invention, comprising mixing the oligonucleotide or conjugated oligonucleotide of the invention with a pharmaceutically acceptable diluent, solvent, carrier, salt and/or adjuvant.
  • Pharmaceutical Salts
  • For use as a therapeutic, the oligonucleotide of the invention may be provided as a suitable pharmaceutical salt, such as a sodium or potassium salt. In some embodiments the oligonucleotide of the invention is a sodium salt.
  • Pharmaceutical Composition
  • In a further aspect, the invention provides pharmaceutical compositions comprising any of the aforementioned oligonucleotides and/or oligonucleotide conjugates and a pharmaceutically acceptable diluent, carrier, salt and/or adjuvant. A pharmaceutically acceptable diluent includes phosphate-buffered saline (PBS) and pharmaceutically acceptable salts include, but are not limited to, sodium and potassium salts. In some embodiments the pharmaceutically acceptable diluent is sterile phosphate buffered saline. In some embodiments the oligonucleotide is used in the pharmaceutically acceptable diluent at a concentration of 50-300 μM solution. In some embodiments, the oligonucleotide of the invention is administered at a dose of 10-1000 μg.
  • WO 2007/031091 provides suitable and preferred examples of pharmaceutically acceptable diluents, carriers and adjuvants (hereby incorporated by reference). Suitable dosages, formulations, administration routes, compositions, dosage forms, combinations with other therapeutic agents, pro-drug formulations are also provided in WO2007/031091.
  • Oligonucleotides or oligonucleotide conjugates of the invention may be mixed with pharmaceutically acceptable active or inert substances for the preparation of pharmaceutical compositions or formulations. Compositions and methods for the formulation of pharmaceutical compositions are dependent upon a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered.
  • In some embodiments, the oligonucleotide or oligonucleotide conjugate of the invention is a prodrug. In particular with respect to oligonucleotide conjugates the conjugate moiety is cleaved of the oligonucleotide once the prodrug is delivered to the site of action, e.g. the target cell.
  • Applications
  • The oligonucleotides of the invention may be utilized as research reagents for, for example, diagnostics, therapeutics and prophylaxis.
  • In research, such oligonucleotides may be used to specifically modulate the synthesis of HTRA1 protein in cells (e.g. in vitro cell cultures) and experimental animals thereby facilitating functional analysis of the target or an appraisal of its usefulness as a target for therapeutic intervention. Typically the target modulation is achieved by degrading or inhibiting the mRNA producing the protein, thereby prevent protein formation or by degrading or inhibiting a modulator of the gene or mRNA producing the protein.
  • In diagnostics the oligonucleotides may be used to detect and quantitate HTRA1 expression in cell and tissues by northern blotting, in-situ hybridisation or similar techniques.
  • For therapeutics, an animal or a human, suspected of having a disease or disorder, which can be treated by modulating the expression of HTRA1.
  • The invention provides methods for treating or preventing a disease, comprising administering a therapeutically or prophylactically effective amount of an oligonucleotide, an oligonucleotide conjugate or a pharmaceutical composition of the invention to a subject suffering from or susceptible to the disease.
  • The invention also relates to an oligonucleotide, a composition or a conjugate as defined herein for use as a medicament.
  • The oligonucleotide, oligonucleotide conjugate or a pharmaceutical composition according to the invention is typically administered in an effective amount.
  • The invention also provides for the use of the oligonucleotide or oligonucleotide conjugate of the invention as described for the manufacture of a medicament for the treatment of a disorder as referred to herein, or for a method of the treatment of as a disorder as referred to herein.
  • The disease or disorder, as referred to herein, is associated with expression of HTRA1. In some embodiments disease or disorder may be associated with a mutation in the HTRA1 gene or a gene whose protein product is associated with or interacts with HTRA1. Therefore, in some embodiments, the target nucleic acid is a mutated form of the HTRA1 sequence and in other embodiments, the target nucleic acid is a regulator of the HTRA1 sequence.
  • The methods of the invention are preferably employed for treatment or prophylaxis against diseases caused by abnormal levels and/or activity of HTRA1.
  • The invention further relates to use of an oligonucleotide, oligonucleotide conjugate or a pharmaceutical composition as defined herein for the manufacture of a medicament for the treatment of abnormal levels and/or activity of HTRA1.
  • In one embodiment, the invention relates to oligonucleotides, oligonucleotide conjugates or pharmaceutical compositions for use in the treatment of diseases or disorders selected from eye disorders, such as macular degeneration, including age related macular degeneration (AMD), such as dry AMD or wet AMD, and diabetic retinopathy. In some embodiments the oligonucleotide conjugates or pharmaceutical compositions of the invention may be for use in the treatment of geographic atrophy or intermediate dAMD. HTRA1 has also been indicated in Alzheimer's and Parkinson's disease, and therefore in some embodiments, the oligonucleotide conjugates or pharmaceutical compositions of the invention may be for use in the treatment of Alzheimer's or Parkinson's. HTRA1 has also been indicated in Duchenne muscular dystrophy, arthritis, such as osteoarthritis, familial ischemic cerebral small-vessel disease, and therefore in some embodiments, the oligonucleotide conjugates or pharmaceutical compositions of the invention may be for use in the treatment of Duchenne muscular dystrophy, arthritis, such as osteoarthritis, or familial ischemic cerebral small-vessel disease.
  • Administration
  • The oligonucleotides or pharmaceutical compositions of the present invention may be administered topical (such as, to the skin, inhalation, ophthalmic or otic) or enteral (such as, orally or through the gastrointestinal tract) or parenteral (such as, intravenous, subcutaneous, intra-muscular, intracerebral, intracerebroventricular or intrathecal).
  • In some embodiments the oligonucleotide, conjugate or pharmaceutical compositions of the present invention are administered by a parenteral route including intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion, intrathecal or intracranial, e.g. intracerebral or intraventricular, administration. In some embodiments the active oligonucleotide or oligonucleotide conjugate is administered intravenously. In another embodiment the active oligonucleotide or oligonucleotide conjugate is administered subcutaneously.
  • For use in treating eye disorders, such as macular degeneration, e.g. AMD (wet or dry), intraocular injection may be used.
  • In some embodiments, the compound of the invention, or pharmaceutically acceptable salt thereof, is administered via an intraocular injection in a dose from about 10 μg to about 200 μg per eye, such as about 50 μg to about 150 μg per eye, such as about 100 μg per eye. In some embodiments, the dosage interval, i.e. the period of time between consecutive dosings is at least monthly, such as at least bi monthly or at least once every three months.
  • Combination Therapies
  • In some embodiments the oligonucleotide, oligonucleotide conjugate or pharmaceutical composition of the invention is for use in a combination treatment with another therapeutic agent. The therapeutic agent can for example be the standard of care for the diseases or disorders described above
  • Embodiments of the Invention
  • 1. An antisense oligonucleotide of 10-30 nucleotides in length, wherein said antisense oligonucleotide comprises a contiguous nucleotide region of 10-22 nucleotides which are at least 90% such as 100% complementarity to SEQ ID NO 147:
  • SEQ ID NO 147: 5′ CCAACAACCAGGTAAATATTTG 3′
  • 2. The antisense oligonucleotide according to embodiment 1, wherein the antisense oligonucleotide is capable of inhibiting the expression of HTRA1 mRNA.
  • 3. The antisense oligonucleotide according to embodiment 1 or 2, wherein the contiguous nucleotide region is identical to a sequence present in a sequence selected from the group consisting of SEQ ID NO 138, 139, 140, 141, 142, 143, 144 and 145:
  • SEQ ID NO 138: CAAATATTTACCTGGTTG
    SEQ ID NO 139: TTTACCTGGTTGTTGG
    SEQ ID NO 140: CCAAATATTTACCTGGTT
    SEQ ID NO 141: CCAAATATTTACCTGGTTGT
    SEQ ID NO 142: ATATTTACCTGGTTGTTG
    SEQ ID NO 143: TATTTACCTGGTTGTT
    SEQ ID NO 144: ATATTTACCTGGTTGT
    SEQ ID NO 145: ATATTTACCTGGTTGTT
  • 4. The antisense oligonucleotide according to any one of embodiments 1-3, wherein the contiguous nucleotide region comprises the sequence SEQ ID NO 146:
  • SEQ ID NO 146: TTTACCTGGTT
  • 5. The antisense oligonucleotide according to any one of embodiments 1-4, wherein the contiguous nucleotide region of the oligonucleotide consists or comprises of a sequence selected from any one of SEQ ID NO 138, 139, 140, 141, 142, 143, 144 and 145.
  • 6. The antisense oligonucleotide according to any one of embodiments 1-5 wherein the contiguous nucleotide region of the oligonucleotide comprises one or more 2′ sugar modified nucleosides.
  • 7. The antisense oligonucleotide according to embodiment 6, wherein the one or more 2′ sugar modified nucleoside is independently selected from the group consisting of 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA, 2′-amino-DNA, 2′-fluoro-DNA, arabino nucleic acid (ANA), 2′-fluoro-ANA and LNA nucleosides.
  • 8. The antisense oligonucleotide according to any one of embodiments 5-7, wherein the one or more modified nucleoside is a LNA nucleoside.
  • 9. The antisense oligonucleotide according to any one of embodiments 1-8, where the contiguous nucleotide region of the oligonucleotide comprises at least one modified internucleoside linkage, such as one or more phosphorothioate internucleoside linkages.
  • 10. The antisense oligonucleotide according to embodiment 9, wherein all the internucleoside linkages within the contiguous nucleotide region are phosphorothioate internucleoside linkages.
  • 11. The antisense oligonucleotide according to any one of embodiments 1-10, wherein the oligonucleotide is capable of recruiting RNase H.
  • 12. The antisense oligonucleotide according to any one of embodiments 1-11, wherein the oligonucleotide or contiguous nucleotide sequence thereof is or comprises a gapmer.
  • 13. The antisense oligonucleotide of embodiment 11 or 12, wherein the oligonucleotide or contiguous nucleotide sequence thereof is a gapmer of formula 5′-F-G-F′-3′, where region F and F′ independently comprise 1-7 sugar modified nucleosides and G is a region 6-16 nucleosides which is capable of recruiting RNaseH, wherein the nucleosides of regions F and F′ which are adjacent to region G are sugar modified nucleosides.
  • 14. The antisense oligonucleotide according to embodiment 13, wherein region G consists or comprises 6-16 DNA nucleosides.
  • 15. The antisense oligonucleotide according to embodiment 13 or 14, wherein region F and F′ each comprise at least one LNA nucleoside.
  • 16. The antisense oligonucleotide according to any one of embodiments 1-15, selected from the group selected from:
  • (SEQ ID NO 138, Comp # 138.1)
    CsAsAsAstsastststsascscstsgsGsTsTsG 
    (SEQ ID NO 139, Comp # 139.1)
    TsTstsascscstsgsgststsgstsTsGsG 
    (SEQ ID NO 140, Comp # 140.1)
    CsCsAsAsastsastststsascscstsgsGsTsT 
    (SEQ ID NO 141, Comp # 141.1)
    CsCsAsasastsastststsascscstsgsgststsGsT 
    (SEQ ID NO 142, Comp # 142.1)
    AsTsAstststsascscstsgsgststsgsTsTsG 
    (SEQ ID NO 143, Comp # 143.1)
    TsAsTststsascscstsgsgstsTsGsTsT 
    (SEQ ID NO 143, Comp # 143.2)
    TsAstststsascscstsgsGstsTsgsTsT 
    (SEQ ID NO 143, Comp # 143.3)
    TsAsTststsascscstsgsgsTsTsgsTsT 
    (SEQ ID NO 144, Comp # 144.1)
    AsTsAsTststsascscstsgsgsTsTsGsT 
    (SEQ ID NO 144, Comp # 144.2)
    AstsAsTsTstsascscstsgsgstsTsGsT 
    (SEQ ID NO 145, Comp # 145.1)
    AstsAsTststsascscstsgsGsTsTsgsTsT 
    (SEQ ID NO 145, Comp # 145.2)
    AsTsAstststsascscstsgsGstsTsgsTsT 
    (SEQ ID NO 145, Comp # 145.3)
    AstsAsTststsascscstsgsgstsTsGsTsT 
  • Wherein a capital letter represents an LNA nucleoside unit, a lower case letter represents a DNA nucleoside unit, subscript s represents a phosphorothioate internucleoside linkage, wherein all LNA cytosines are 5-methyl cytosine.
  • 17. The antisense oligonucleotide according to embodiment 16, wherein the LNA nucleosides are all beta-D-oxy LNA nucleosides.
  • 18. A conjugate comprising the oligonucleotide according to any one of embodiments 1-17, and at least one conjugate moiety covalently attached to said oligonucleotide.
  • 19. A pharmaceutical composition comprising the oligonucleotide of embodiment 1-17 or the conjugate of embodiment 18 and a pharmaceutically acceptable diluent, solvent, carrier, salt and/or adjuvant.
  • 20. An in vivo or in vitro method for modulating HTRA1 expression in a target cell which is expressing HTRA1, said method comprising administering an oligonucleotide of any one of embodiments 1-17 or the conjugate according to embodiment 18 or the pharmaceutical composition of embodiment 19 in an effective amount to said cell.
  • 21. A method for treating or preventing a disease comprising administering a therapeutically or prophylactically effective amount of an oligonucleotide of any one of embodiments 1-17 or the conjugate according to embodiment 18 or the pharmaceutical composition of embodiment 19 to a subject suffering from or susceptible to the disease.
  • 22. The method of embodiment 21, wherein the disease is selected from the group consisting of macular degeneration (such as wetAMD, dryAMD, geographic atrophy, intermediate dAMD, diabetic retinopathy), Parkinson's disease, Alzhiemer's disease, Duchenne muscular dystrophy, arthritis, such as osteoarthritis, and familial ischemic cerebral small-vessel disease.
  • 23. The oligonucleotide of any one of embodiments 1-17 or the conjugate according to embodiment 18 or the pharmaceutical composition of embodiment 19 for use in medicine.
  • 24. The oligonucleotide of any one of embodiments 1-17 or the conjugate according to embodiment 18 or the pharmaceutical composition of embodiment 19 for use in the treatment or prevention of a disease is selected from the group consisting of macular degeneration (such as wetAMD, dryAMD, geographic atrophy, intermediate dAMD, diabetic retinopathy), Parkinson's disease, Alzhiemer's disease, Duchenne muscular dystrophy, arthritis, such as osteoarthritis, and familial ischemic cerebral small-vessel disease.
  • 25. Use of the oligonucleotide of embodiment 1-17 or the conjugate according to embodiment 18 or the pharmaceutical composition of embodiment 19, for the preparation of a medicament for treatment or prevention of a disease is selected from the group consisting of macular degeneration (such as wetAMD, dryAMD, geographic atrophy, intermediate dAMD, diabetic retinopathy), Parkinson's disease, Alzhiemer's disease, Duchenne muscular dystrophy, arthritis, such as osteoarthritis, and familial ischemic cerebral small-vessel disease.
  • Examples
  • Materials and Methods
  • Oligonucleotide Synthesis
  • Oligonucleotide synthesis is generally known in the art. Below is a protocol which may be applied. The oligonucleotides of the present invention may have been produced by slightly varying methods in terms of apparatus, support and concentrations used.
  • Oligonucleotides are synthesized on uridine universal supports using the phosphoramidite approach on an Oligomaker 48 at 1 μmol scale. At the end of the synthesis, the oligonucleotides are cleaved from the solid support using aqueous ammonia for 5-16 hours at 60° C. The oligonucleotides are purified by reverse phase HPLC (RP-HPLC) or by solid phase extractions and characterized by UPLC, and the molecular mass is further confirmed by ESI-MS.
  • Elongation of the Oligonucleotide:
  • The coupling of β-cyanoethyl-phosphoramidites (DNA-A(Bz), DNA-G(ibu), DNA-C(Bz), DNA-T, LNA-5-methyl-C(Bz), LNA-A(Bz), LNA-G(dmf), LNA-T) is performed by using a solution of 0.1 M of the 5′-O-DMT-protected amidite in acetonitrile and DCI (4,5-dicyanoimidazole) in acetonitrile (0.25 M) as activator. For the final cycle a phosphoramidite with desired modifications can be used, e.g. a C6 linker for attaching a conjugate group or a conjugate group as such. Thiolation for introduction of phosphorthioate linkages is carried out by using xanthane hydride (0.01 M in acetonitrile/pyridine 9:1). Phosphordiester linkages can be introduced using 0.02 M iodine in THF/Pyridine/water 7:2:1. The rest of the reagents are the ones typically used for oligonucleotide synthesis.
  • For post solid phase synthesis conjugation a commercially available C6 aminolinker phorphoramidite can be used in the last cycle of the solid phase synthesis and after deprotection and cleavage from the solid support the aminolinked deprotected oligonucleotide is isolated. The conjugates are introduced via activation of the functional group using standard synthesis methods.
  • Purification by RP-HPLC:
  • The crude compounds are purified by preparative RP-HPLC on a Phenomenex Jupiter C18 10μ 150×10 mm column. 0.1 M ammonium acetate pH 8 and acetonitrile is used as buffers at a flow rate of 5 mL/min. The collected fractions are lyophilized to give the purified compound typically as a white solid.
  • Abbreviations
  • DCI: 4,5-Dicyanoimidazole
  • DCM: Dichloromethane
  • DMF: Dimethylformamide
  • DMT: 4,4′-Dimethoxytrityl
  • THF: Tetrahydrofurane
  • Bz: Benzoyl
  • Ibu: Isobutyryl
  • RP-HPLC: Reverse phase high performance liquid chromatography
  • Tm Assay:
  • Oligonucleotide and RNA target (phosphate linked, PO) duplexes are diluted to 3 mM in 500 ml RNase-free water and mixed with 500 ml 2×Tm-buffer (200 mM NaCl, 0.2 mM EDTA, 20 mM Naphosphate, pH 7.0). The solution is heated to 95° C. for 3 min and then allowed to anneal in room temperature for 30 min. The duplex melting temperatures (Tm) is measured on a Lambda 40 UV/VIS Spectrophotometer equipped with a Peltier temperature programmer PTP6 using PE Templab software (Perkin Elmer). The temperature is ramped up from 20° C. to 95° C. and then down to 25° C., recording absorption at 260 nm. First derivative and the local maximums of both the melting and annealing are used to assess the duplex Tm.
  • Example 1: Testing In Vitro Efficacy of Antisense Oligonucleotides Targeting Rat Htra1 in C6 Cell Lines at Single Dose Concentration
  • Rat C6 cell line was purchased from ATCC and maintained as recommended by the supplier in a humidified incubator at 37° C. with 5% CO2. For assays, 1500 C6 cells/well were seeded in a 96 multi well plate in culture media. Cells were incubated for 2 hours before addition of oligonucleotides dissolved in PBS. Concentration of oligonucleotides: 25 μM. 4 days after addition of oligonucleotides, the cells were harvested. RNA was extracted using the PureLink Pro 96 RNA Purification kit (Ambion, according to the manufacturer's instructions). cDNA was then synthesized using M-MLT Reverse Transcriptase, random decamers RETROscript, RNase inhibitor (Ambion, according the manufacturer's instruction) with 100 mM dNTP set PCR Grade (Invitrogen) and DNase/RNase free Water (Gibco). For gene expressions analysis, qPCR was performed using TagMan Fast Advanced Master Mix (2×) (Ambion) in a doublex set up. Following TaqMan primer assays were used for qPCR: Htra1, Rn00581870_m1 (FAM-MGB) and house keeping gene, Tbp, Rn01455646_m1 (VIC-MGB). All primer sets were purchase from Life Technologies. The relative Htra1 mRNA expression level in the table is shown as % of control (PBS-treated cells).
  • Oligonucleotides Used:
  • SEQ ID NO Motif CMP ID NO Compound mRNA level
    5 gaaagggaaatatggg 5.1 GAAagggaaatatGGG 51
    6 gatgaggtataaagtg 6.1 GATgaggtataaaGTG 54
    7 ggtgtgttaataatca 7.1 GGTgtgttaataaTCA 60
    8 cttatgacgcaaactg 8.1 CTTatgacgcaaaCTG 26
    9 tttgtctcctttcctc 9.1 TTtgtctcctttccTC 70
    10 gaatggaaagatgtaa 10.1 GAATggaaagatGTAA 27
    11 gttctttggctttgct 11.1 GTtctttggctttgCT 63
    12 ttcaatgatatatgct 12.1 TTCaatgatatatGCT 15
    13 agtatgaagaagtatt 13.1 AGTatgaagaagtATT 27
    14 cccaatcacctcgcca 14.1 CCCaatcacctmcgCCA 72
    15 gcagtagcaaagacagg 15.1 GCagtagcaaagacAGG 28
    16 aagttgaaatcagtggt 16.1 AAGttgaaatcagTGGT 12
    17 tctggtagtaagaatata 17.1 TCTGgtagtaagaaTATA 73
    18 aacagtaagagctacttt 18.1 AACAgtaagagctaCTTT 62
    19 tcagacacgatacagag 19.1 TCAgacamcgatacAGAG 39
    20 tggtcagtgataagtaa 20.1 TGGtcagtgataaGTAA 34
    21 gcactgtagatgagaaac 21.1 GCACtgtagatgagAAAC 37
    22 ataaagtaaacttaatgcc 22.1 ATAAagtaaacttaaTGCC 32
    23 attggttcttaggagtgggc 23.1 AttggttcttaggagtggGC 56
    24 attattgttttactcgtga 24.1 ATTattgttttactmcgTGA 32
    25 atgctggggtaatgattg 25.1 ATGctggggtaatgaTTG 58
    26 agtctaaattattgcacaa 26.1 AGTCtaaattattgcACAA 72
    27 ccaattagaacagtagtgg 27.1 CCAAttagaacagtagtGG 58
    28 agtgtctagttaaacagcac 28.1 AGtgtctagttaaacagCAC 84
    29 taccagagtcaagcatatg 29.1 TACcagagtcaagcataTG 33
    30 atctaaacttcatgtcagaa 30.1 ATCtaaacttcatgtcAGAA 86
    31 ttcatacgactgagcatc 31.1 TTCAtamcgactgagcATC 26
    32 gtacagttttagatcatc 32.1 GTAcagttttagatCATC 18
    33 atcctaaaagagttcaaca 33.1 ATCCtaaaagagttcaACA 77
    34 cattctttgtcatatact 34.1 CATtctttgtcataTACT 49
    35 ttcaaagtgactttcaag 35.1 TTCAaagtgactttCAAG 11
    36 tcctttctataattacaaa 36.1 TCCTttctataattaCAAA 35
    37 ctaattctgtagttttgg 37.1 CTaattctgtagttTTGG 31
    38 actgagtgtgtaatgttga 38.1 ACtgagtgtgtaatgtTGA 59
    39 agagaaatctgtagggc 39.1 AGAgaaatctgtaggGC 56
    40 ttatcaaatcaactagcac 40.1 TTAtcaaatcaactaGCAC 84
    41 ttatatttgatagtgtgatc 41.1 TTATatttgatagtgtgATC 55
    42 aagagaagctgttatctaaa 42.1 AAGAgaagctgttatcTAAA 42
    43 aagtcatgctagagttcc 43.1 AAgtcatgctagagtTCC 37
    44 acacagtgtattcgaggg 44.1 ACAcagtgtattmcgagGG 38
    45 gacacagtgtattcgaggg 45.1 GAcacagtgtattmcgagGG 52
    46 gacacagtgtattcgagg 46.1 GACacagtgtattmcgaGG 26
    47 aggacacagtgtattcgagg 47.1 AGgacacagtgtattmcgaGG 38
    48 aggacacagtgtattcgag 48.1 AGgacacagtgtattmcgAG 32
    49 caggacacagtgtattcgag 49.1 CAggacacagtgtattmcgAG 53
    50 aggacacagtgtattcg 50.1 AGgacacagtgtaTTCG 11
    51 aacaggacacagtgtattcg 51.1 AAcaggacacagtgtaTTCG 28
    52 tgaatctatacagcaggaa 52.1 TGAAtctatacagcagGAA 46
    53 cttaagttattcatatcca 53.1 CTTaagttattcatatCCA 39
    54 taggtggtagcagatag 54.1 TAGgtggtagcagatAG 48
    55 gtagtaataattctggga 55.1 GTAgtaataattctgGGA 7
    56 agtggtaaggtgaagtgaa 56.1 AGtggtaaggtgaagTGAA 45
    57 ttttgctgtgataaatagc 57.1 TTttgctgtgataaaTAGC 65
    58 tttcttatcgttttatga 58.1 TTTCttatmcgttttATGA 24
    59 ggaaaacattaacaaggttg 59.1 GGAAaacattaacaagGTTG 38
    60 tgggtagagtctaggag 60.1 TGggtagagtctaggAG 58
    61 agggagtgactattagag 61.1 AGggagtgactattaGAG 45
    62 cagagtagggaaagtggttc 62.1 CAgagtagggaaagtggtTC 76
    63 accatgttatatttggg 63.1 ACCAtgttatatttgGG 54
    64 tttattactgaggggaaagg 64.1 TTTAttactgaggggaaaGG 67
    65 acttgagtgtagtacag 65.1 ACTtgagtgtagtACAG 21
    66 catactagttttgcagaga 66.1 CAtactagttttgcagAGA 54
    67 ggactagaagtagttac 67.1 GGActagaagtagTTAC 58
    68 agggagtcaaaggttcaa 68.1 AGggagtcaaaggtTCAA 13
    69 tctgtgtattagagaacg 69.1 TCTGtgtattagagAACG 44
    70 tcaaaagctacatcagtc 70.1 TCAaaagctacatcAGTC 7
    71 accgttgaattagtcac 71.1 ACCGttgaattagtCAC 39
    72 tttccattaaaatgtttaca 72.1 TTTCcattaaaatgttTACA 28
    73 gagatggtaaggagtaggag 73.1 GAgatggtaaggagtaggAG 60
    74 agttttagactattctg 74.1 AGTTttagactattCTG 37
    75 gtcaggacataaactcac 75.1 GTCaggacataaacTCAC 18
    76 ttgttggtgtcagggaaaag 76.1 TTGttggtgtcagggaaaAG 20
    77 tcctgaaatattgatgc 77.1 TCCtgaaatattgaTGC 51
    78 tgccaaaatgactacagt 78.1 TGCcaaaatgactacAGT 54
    79 ctcaaagttggatcgtaac 79.1 CTCaaagttggatmcgTAAC 37
    80 ttgcattttagaagttat 80.1 TTGCattttagaagTTAT 49
    81 tgtcaattagtgtgtttag 81.1 TGTCaattagtgtgtttAG 20
    82 cataatatttagtctcct 82.1 CATaatatttagtctCCT 40
    83 tatagttacaatcacca 83.1 TATAgttacaatcaCCA 30
    84 ttttgtacatactcttcc 84.1 TTttgtacatactcTTCC 11
    85 tagcagtacttaaatggg 85.1 TAGcagtacttaaatGGG 28
    86 tatgttacatatttggatga 86.1 TATgttacatatttggaTGA 41
    87 catggtatcttttggagag 87.1 CATggtatcttttggagAG 36
    88 caacatatccagtccag 88.1 CAAcatatccagtcCAG 22
    89 taattgtgaagtagggtg 89.1 TAattgtgaagtagGGTG 33
    90 actttagaggtttagtcc 90.1 ACTttagaggtttagtCC 55
    91 gagcttttaattctatcc 91.1 GAgcttttaattctATCC 56
    92 ctccttaaatacatgttac 92.1 CTCcttaaatacatgTTAC 49
    93 gagataaatgtttgagaga 93.1 GAGAtaaatgtttgagAGA 41
    94 atgatggtgatttaggat 94.1 ATGAtggtgatttagGAT 22
    95 agtcattcaatttgagaaa 95.1 AGTCattcaatttgaGAAA 35
    96 cagtggtggtaaggcac 96.1 CAGtggtggtaaggcAC 53
    97 tgaaatggtttagttctg 97.1 TGAAatggtttagttCTG 41
    98 tgccatagtgaaatggttt 98.1 TGCcatagtgaaatggtTT 57
    99 caggaggacatactatt 99.1 CAGgaggacatacTATT 77
    100 atatatacaggcacatgg 100.1 ATAtatacaggcacaTGG 71
    101 gacaggagtctttaaaatg 101.1 GACAggagtctttaaAATG 80
    102 tatttatatagtaatgtgtc 102.1 TATTtatatagtaatgTGTC 57
    103 ggataaaacagtaccat 103.1 GGAtaaaacagtaCCAT 77
    104 ggagtttagaagacacat 104.1 GGAgtttagaagacaCAT 33
    105 gtacaactacagaggtt 105.1 GTACaactacagagGTT 65
    106 aggaaatggtgatggaatg 106.1 AGGaaatggtgatggAATG 58
    107 tcggagtaaaagtgtaaaca 107.1 TCGGagtaaaagtgtaaACA 81
    For Compounds: Capital letters represent LNA nucleosides (beta-D-oxy LNA nucleosides were used), all LNA cytosines are 5-methyl cytosine, lower case letters represent DNA nucleosides, DNA cytosines preceded with a superscript m represents a 5-methyl C-DNA nucleoside. All internucleoside linkages are phosphorothioate internucleoside linkages.
  • Example 2. Testing In Vitro Potency and Efficacy of Selected Oligonucleotides Targeting Rat Htra1 in C6 Cell Line in a Dose Response Curve
  • Rat C6 cell line was described in Example 1. The assay was performed as described in Example 1. Concentration of oligonucleotides: from 50 μM, half-log dilution, 8 points. 4 days after addition of oligonucleotides, the cells were harvested. RNA extraction, cDNA synthesis and qPCR were performed as described in Example 1. n=2 biological replicates. EC50 determinations were performed in GraphPad Prism6. The relative Htra1 mRNA level at treatment with 50 μM oligonucleotide is shown in the table as % of control (PBS). Additional primer sets (Htra1, Rn00668987_m1 [FAM-MGB] vs. Ppia, Rn006900933_m1 [VIC-MGB] and Hprt, Rn01527840_m1 [VIC-MGB]) were also tested and the same trends were observed using those primers (data not shown).
  • mRNA level
    SEQ ID NO CMP ID NO EC50 at Max KD
    12 12.1 3.3 14
    16 16.1 3.2 11
    32 32.1 4.3 27
    35 35.1 2.6 8
    50 50.1 1.9 9
    55 55.1 2.0 6
    58 58.1 4.4 25
    65 65.1 3.1 14
    68 68.1 4.1 8
    70 70.1 1.5 2
    75 75.1 3.7 16
    76 76.1 4.9 21
    81 81.1 3.0 18
    84 84.1 2.2 9
    88 88.1 6.8 20
    94 94.1 2.9 15
  • Example 3 Testing In Vitro Efficacy of Oligonucleotides Targeting Human HTRA1, in U251 Cell Line at Single Dose Concentration
  • Human glioblastoma U251 cell line was purchased from ECACC and maintained as recommended by the supplier in a humidified incubator at 37° C. with 5% CO2. For assays, 15000 U251 cells/well were seeded in a 96 multi well plate in starvation media (media recommended by the supplier with the exception of 1% FBS instead of 10%). Cells were incubated for 24 hours before addition of oligonucleotides dissolved in PBS. Concentration of oligonucleotides: 5 μM. 3-4 days after addition of compounds, media was removed and new media (without oligonucleotide) was added. 6 days after addition of oligonucleotides, the cells were harvested. RNA was extracted using the PureLink Pro 96 RNA Purification kit (Ambion, according to the manufacturer's instructions). cDNA was then synthesized using M-MLT Reverse Transcriptase, random decamers RETROscript, RNase inhibitor (Ambion, according the manufacturer's instruction) with 100 mM dNTP set PCR Grade (Invitrogen) and DNase/RNase free Water (Gibco). For gene expressions analysis, qPCR was performed using TagMan Fast Advanced Master Mix (2×) (Ambion) in a doublex set up. Following TaqMan primer assays were used for qPCR: HTRA1, Hs01016151_m1 (FAM-MGB) and house keeping gene, TBP, Hs4326322E (VIC-MGB) from Life Technologies. The relative HTRA1 mRNA expression level in the table is shown as % of control (PBS-treated cells).
  • SEQ ID NO Motif CMP ID NO Compound mRNA level
    108 agatgggtgtgaaagg 108.1 AGAtgggtgtgaaAGG 35
    109 atgttgtctatgttta 109.1 ATGttgtctatgtTTA 5
    110 tatgttgtctatgttt 110.1 TATgttgtctatgTTT 5
    111 gatgtttgcagtattt 111.1 GATgtttgcagtaTTT 17
    112 tatatagtcgaatagg 112.1 TATatagtcgaatAGG 18
    113 tttggcttcgtaagtg 113.1 TTTggcttcgtaaGTG 1
    114 tgaggcagtggagttg 114.1 TGaggcagtggagtTG 32
    115 tggacaggagggcagc 115.1 TGgacaggagggcaGC 92
    116 tagagaaggtagaatg 116.1 TAGagaaggtagaATG 78
    117 atttagattagagaag 117.1 ATTTagattagaGAAG 50
    118 gttcttaaatgtcgtt 118.1 GTTcttaaatgtcGTT 10
    119 aagggcttaccatctt 119.1 AAGggcttaccatCTT 62
    120 tacttcaattatatac 120.1 TACttcaattataTAC 25
    121 gcaatgtgtaagaagt 121.1 GCAatgtgtaagaAGT 15
    122 aaactgttgggatctt 122.1 AAACtgttgggaTCTT 11
    123 caaactgttgggatct 123.1 CAAActgttgggATCT 50
    124 gcaaactgttgggatc 124.1 GCAaactgttgggATC 11
    125 gatgtttgcagtattt 125.1 GAtgtttgcagtaTTT 22
    126 attgggtttgatcggt 126.1 ATTgggtttgatmcgGT 15
    127 ctattgggtttgatcg 127.1 CTAttgggtttgatCG 16
    128 tattgggtttgatcgg 128.1 TATtgggtttgatCGG 10
    129 cgaatatgtgctttaa 129.1 CGAatatgtgcttTAA 8
    130 gctgattatgacgtcg 130.1 GCTgattatgamcgTCG 13
    131 tgctgattatgacgtc 131.1 TGCtgattatgamcGTC 10
    132 attgggtttgatcggt 132.1 ATTgggtttgatCGGT 18
    133 ctattgggtttgatcg 133.1 CTAttgggtttgATCG 49
    134 tgctgattatgacgtc 134.1 TGCtgattatgaCGTC 46
    135 tattgggtttgatcgg 135.1 TATTgggtttgaTCGG 58
    136 cgaatatgtgctttaa 136.1 CGAAtatgtgctTTAA 13
    137 gctgattatgacgtcg 137.1 GCTGattatgamcGTCG 39
    For Compounds: Capital letters represent LNA nucleosides (beta-D-oxy LNA nucleosides were used), all LNA cytosines are 5-methyl cytosine, lower case letters represent DNA nucleosides, DNA cytosines preceded with a superscript m represents a 5-methyl C-DNA nucleoside. All internucleoside linkages are phosphorothioate internucleoside linkages.
  • Example 4 Testing In Vitro Efficacy of a Library of Oligonucleotides Targeting Human HTRA1 mRNA in ARPE19 and U251 Cell Lines at 2 Concentrations
  • Identification of promising “hot spot” region for HTRA1. A library of n=129 human/cyno/rat HTRA1 LNA oligonucleotides were screened in U251 and ARPE19 cell lines. From this library we identified a series of active oligonucleotides targeting human HTRA1 pre-mRNA (SEQ ID NO 2) between position 33042-33064 as shown in FIG. 1.
  • Human retinal pigmented epithelium ARPE19 cell line was purchased by from ATCC and maintained in DMEM-F12 (Sigma, D8437), 10% FBS, 1% pen/strep in a humidified incubator at 37° C. with 5% CO2. The U251 cell line was described in example 3. For assays, 5000 ARPE19 cells/well were seeded in a 96 multi well plate in culture media (with the exception of 5% FBS instead of 10%). Cells were incubated for 1 hour before addition of oligonucleotides dissolved in PBS. 4 days after addition of oligonucleotides, the cells were harvested. The assay with the U251 cell line was performed as described in example 3. Concentration of oligonucleotides: 25 and 2.5 μM. RNA was extracted using the RNeasy 96 Biorobot 8000 kit (Qiagen, according to the manufacturer's instructions). cDNA was then synthesized using Retroscript cDNA synthesis kit (ThermoFisher, according the manufacturer's instruction). For gene expressions analysis, qPCR was performed using the Fluidigm Biomark system. Following TaqMan primer assays were used for qPCR: HTRA1, Hs01016151_m1 and house-keeping genes, TBP, Hs99999910_m1 and PPIA, Hs99999904_m1, from Life Technologies. n=2 biological replicates. The relative HTRA1 mRNA expression level is shown in the table as % of control (PBS). Additional HTRA1 primer set (Hs00170197_m1) was also tested and the same trends were observed (data not shown).
  • ARPE19 U251 mRNA
    SEQ mRNA level level
    ID Comp
    25 2.5 25 2.5
    NO # Compound μM μM μM μM
    138 138.1 mCAAAtatttacctgGTTG 79 97 11 60
    139 139.1 TTtacctggttgaGG 39 73 5 37
    140 140.1 mCmCAAatatttacctgGTT 68 100 16 70
    141 141.1 mCmCAaatatttacctggtt 78 87 16 78
    GT
    142 142.1 ATAtttacctggttgTTG 56 78 4 23
    143 143.1 TATttacctggaGTT 22 77 3 23
    For Compounds: Capital letters represent LNA nucleosides (beta-D-oxy LNA nucleosides were used), all LNA cytosines are 5-methyl cytosine, lower case letters represent DNA nucleosides. All internucleoside linkages are phosphorothioate internucleoside linkages.
  • Example 5, Testing In Vitro Efficacy of Selected Human/Rat HTRA1 Targeting LNA Oligonucleotidesin Rat C6 Cell Lines at Single Dose Concentration
  • Rat C6 cell line was described in Example 1. The assay was performed as described in Example 1. Concentration of oligonucleotides: 25 μM. n=2 biological replicates. The relative Htra1 mRNA expression level in the table is shown as % of control (PBS-treated cells).
  • SEQ ID NO CMP ID NO mRNA level
    139 139.1 24
    140 140.1 6
    142 142.1 26
    143 143.1 16
  • Example 6, Testing In Vitro Potency and Efficacy of Selected Human/Cyno/Rat LNA Oligonucleotides in ARPE19, U251 and C6 Cell Lines in a Dose Response
  • ARPE19, U251 and C6 cell lines were described in example 4, 3 and 1, respectively. For assays, 2000 U251 or ARPE19 cells/well were seeded in a 96 multi well plate in culture media recommended by the supplier. Cells were incubated for 2 hours before addition of oligonucleotides dissolved in PBS. The C6 cell line assay was performed as described in example 1-2. Concentration of oligonucleotides: from 50 μM, half-log dilution, 8 points. 4 days after addition of oligonucleotides, the cells were harvested. RNA extraction, cDNA synthesis and qPCR were performed for all cell lines as described in Example 1. Following TaqMan primer assays were used for U251 and ARPE19 cells: HTRA1, Hs01016151_m1 (FAM-MGB) and house-keeping gene, TBP, Hs4326322E (VIC-MGB). All primer sets were purchased from Life Technologies. n=2 biological replicates. EC50 determinations were performed in Graph Pad Prism6. The relative HTRA1 mRNA level at treatment with 50 μM oligonucleotide is shown in the table as % of control (PBS).
  • ARPE19 U251 C6
    SEQ CMP mRNA mRNA mRNA
    ID ID EC50 level at EC50 level at EC50 level at
    NO NO (μM) max KD (μM) max KD (μM) max KD
    138 138.1 10 63 6.2 36 ND ND
    139 139.1 10 38 3.2 18 ND ND
    140 140.1 7.9 57 4.5 29 1.3 2
    141 141.1 9.3 64 4.5 44 ND ND
    142 142.1 5.8 40 3.9 25 ND ND
    143 143.1 3.3 25 1.7 6.0 3.5 5
  • Example 7, Testing In Vitro Efficacy of Selected Human HTRA1 Targeting Oligonucleotides in ARPE19 and U251 Cell Lines at Single Dose Concentration
  • ARPE19 and U251 cell lines and assays were described in example 6. RNA extraction was performed as described in example 1, cDNA synthesis and qPCR were performed using qScript XLT one-step RT-qPCR ToughMix Low ROX, 95134-100 (Quanta Biosciences). Following TaqMan primer assays were used for U251 and ARPE19 cells in a douplex set up: HTRA1, Hs01016151_m1 (FAM-MGB) and house-keeping gene, GAPDH, Hs4310884E (VIC-MGB). All primer sets were purchased from Life Technologies. n=1 biological replicate. The relative HTRA1 mRNA expression level in the table is shown as % of control (PBS-treated cells). Additional primer sets (HTRA1, Hs00170197_m1 [FAM-MGB] vs. TBP Hs4326322E [VIC-MGB]) were also tested for U251 and the same trends were observed using those primers (data not shown). See FIG. 2.
  • SEQ ID NO CMP ID NO ARPE19 mRNA level U251 mRNA level
    143 143.1 46 7
    143 143.2 49 10
    143 143.3 50 11
    144 144.1 44 19
    144 144.2 16 9
    145 145.1 51 24
    145 145.2 40 9
    145 145.3 42 4
  • Example 8, Testing In Vitro Efficacy and Potency in Human Primary RPE Cells
  • Human primary Retinal Pigmented Epithelium (hpRPE) cells are purchased from Sciencell (Cat #6540). For assays, 5000 hpRPE cells/well are seeded in a Laminin (Laminin 521, BioLamina Cat #LN521-03) coated 96 multi well plate in culture media (EpiCM, Sciencell Cat #4101). They are expanded with this media for one week and differentiated using the following media for 2 weeks: MEM Alpha media (Sigma Cat #M-4526) supplemented with N1 supplement (Sigma Cat #N-6530), Glutamine-Penicillin-Streptomycin (Sigma Cat #G-1146), Non-Essential Amino Acid (NEAA, Sigma Cat #M-7145), Taurine (Sigma Cat #T-0625), Hydrocortisone (Sigma Cat #H-03966), Triiodo-thyronin (Sigma Cat #T-5516) and Bovine Serum Albumin (BSA, Sigma Cat #A-9647). Cells are cultured in a humidified incubator at 37° C. with 5% CO2.
  • On the day of the experiment, cells are incubated for 1 hour with fresh differentiation media before addition of oligonucleotides. These are dissolved in PBS and applied on cells at day 0 and day 4. On day 7, cells are harvested with 50 μl of RLT buffer with β-mercapto-ethanol (Qiagen Cat #79216). The extraction of the RNA is performed according to the user's manual of the Qiagen RNeasy Mini Kit (Cat #74104; Lot 151048073) including DNase I treatment (Cat #79254; Lot 151042674). RNA quality control is performed with the Agilent Bioanalyzer Nano Kit (Agilent; Cat #5067-1511; Lot 1446). Reverse transcription of total RNA into cDNA (cDNA synthesis) is performed using the High Capacity cDNA Reverse Transcription Kit which is based on random hexamer oligonucleotides, according to the manufacturer's instructions (Thermo Fisher Scientific, Cat #4368814; Lot 00314158). The measurement of the cDNA samples is carried out in triplicates, in a 384-well plate format on the 7900HT real-time PCR instrument (Thermo Fisher Scientific). The following TaqMan primer assays are used for qPCR: HTRA1, Hs01016151_m1 and Hs00170197_m1, housekeeping genes, GAPDH, Hs99999905_m1 and PPIA, Hs99999904_m1, from Life Technologies. n=3 biological replicates. The relative HTRA1 mRNA expression level is shown in the table as % of control (PBS). See FIG. 3.
  • Example 9. Rat In Vivo Efficacy Study, 7 Days of Treatment, Intravitreal (IVT) Injection, 30 μg/Eye
  • Animals
  • Experiment was performed on pigmented male Brown Norway rats. Five animals were included in each group of the study, 15 in total.
  • Compounds and Dosing Procedures
  • To start the experiment, the animals were anesthetized with isoflurane, eyes were disinfected and dilated before an intravitreal injection of 30 μg (in 3 μl) per eye.
  • Euthanasia
  • At the end of the in-life phase (Day 7) all rats were euthanized with CO2 before eyes were harvested for dissection. Retina, sclera and vitreous fluid were taken for further analysis.
  • Quantification of HTRA1 RNA Expression
  • Retina samples were dissected. Rat retina snap frozen tissue was kept frozen and was lysed in the testing facility in RLT lysis buffer (Qiagen RNeasy Mini Kit) and RNA extraction was continued according to the user's manual of the Qiagen RNeasy Mini Kit (Cat #74104; Lot 151039852) including DNase I treatment (Cat #79254; Lot 151048613). RNA quality control was performed with the Agilent Bioanalyzer Nano Kit (Agilent; Cat #5067-1511; Lot 1446). Reverse transcription of total RNA into cDNA (cDNA synthesis) was performed using the High Capacity cDNA Reverse Transcription Kit which is based on random hexamer oligonucleotides, according to the manufacturer's instructions (Thermo Fisher Scientific, Cat #4368814, Lot 00314158). The measurement of the cDNA samples was carried out in triplicates, in a 384-well plate format on the 7900HT real-time PCR instrument (Thermo Fisher Scientific). Following TaqMan primer assays were used for qPCR: Htra1, Rn00581870_m1 and housekeeping genes, Gapdh, Rn01775763_g1 and Tbp, Rn01455646_m1, from Life Technologies. Rats/group: 5, n=10 eyes. Each eye was treated as an individual sample. The relative Htra1 mRNA expression level is shown as % of control (PBS). See FIG. 4.
  • Example 10 Rat In Vivo Efficacy Study, 7 Days of Treatment, Intravitreal (IVT) Injection, Dose Response
  • Animals
  • All experiments were performed on pigmented Brown Norway rats. 17 animals were included in each group of the study, 34 in total.
  • Compounds and Dosing Procedures
  • The animals were anesthetized with an intramuscular injection of a mix of xylazine and ketamine. The test item and negative control (PBS) were administered intravitreally in both eyes of anesthetized animals (3 μL per administration) on study day 1.
  • Euthanasia
  • At the end of the in-life phase (Day 8) were euthanized by intraperitoneal an overdose injection of pentobarbital.
  • Oligo Content Measurement and Quantification of Htra1 RNA Expression
  • Both eyeballs of all animals in low-dose and mid-dose group as well as from 5 first animals from high-dose and PBS groups were used for bioanalysis. Immediately after euthanasia, Vitreous (V), Retina (R) and Choroid (CH) were quickly and carefully dissected out on ice and stored at −80° C. until shipment. Retina sample was lysed in 700 μL MagNa Pure 96 LC RNA Isolation Tissue buffer and homogenized by adding 1 stainless steel bead per 2 ml tube 2×1.5 min using a precellys evolution homogenizer followed by 30 min incubation at RT. The samples were centrifuges, 13000 rpm, 5 min. half was set aside for bioanalysis and for the other half, RNA extraction was continued directly
  • For bioanalysis, the samples were diluted 10-50 fold for oligo content measurements with a hybridization ELISA method. A biotinylated LNA-capture probe and a digoxigenin-conjugated LNA-detection probe (both 35 nM in 5×SSCT, each complementary to one end of the LNA oligonucleotide to be detected) was mixed with the diluted homogenates or relevant standards, incubated for 30 minutes at RT and then added to a streptavidine-coated ELISA plates (Nunc cat. no. 436014).
  • The plates were incubated for 1 hour at RT, washed in 2×SSCT (300 mM sodium chloride, 30 mM sodium citrate and 0.05% v/v Tween-20, pH 7.0) The captured LNA duplexes were detected using an anti-DIG antibodies conjugated with alkaline phosphatase (Roche Applied Science cat. No. 11093274910) and an alkaline phosphatase substrate system (Blue Phos substrate, KPL product code 50-88-00). The amount of oligo complexes was measured as absorbance at 615 nm on a Biotek reader.
  • For RNA extraction, cellular RNA large volume kit (05467535001, Roche) was used in the MagNA Pure 96 system with the program: Tissue FF standard LV3.1 according to the instructions of the manufacturer, including DNAse treatment. RNA quality control and concentration were measured with an Eon reader (Biotek). The RNA concentration was normalized across samples, and subsequent cDNA synthesis and qPCR was performed in a one-step reaction using qScript XLT one-step RT-qPCR ToughMix Low ROX, 95134-100 (Quanta Biosciences). The following TaqMan primer assays were used in a duplex reaction: Htra1, Rn00581870_m1 and Rn00668987_m1 and housekeeping genes, HPRT, Rn01527840_m1 and Tbp, Rn01455646_m1, from Life Technologies. The qPCR analyses were run on a ViiA7 machine (Life Technologies). Rats/group: 5, n=10 eyes. Each eye was treated as an individual sample. The relative Htra1 mRNA expression level is shown as % of control (PBS).
  • Histology
  • Both eyeballs of the 2 remaining animals of high dose and PBS animals were removed and fixed in 10% neutral buffered formalin for 24 hours, trimmed and embedded in paraffin. For ISH analysis, sections of formalin-fixed, paraffin-embedded rat retina tissue 4 um thick were processed using the fully automated Ventana Dicovery ULTRA Staining Module (Procedure: mRNA Discovery Ultra Red 4.0-v0.00.0152) using the RNAscope 2.5 VS Probe-Rn-HTRA1 (Cat No. 440959, Advanced Cell Diagnostic). Chromogen used is Fasted, Hematoxylin H counterstain.
  • Example 11
  • PoC study, Blue Light-induced retinal degeneration in albino rats
  • Animals
  • All experiments were performed on albino Sprague-Dawley rats. Sixteen animals were included in each group of the study, 42 in total.
  • Compounds and Dosing Procedures
  • The animals were anesthetized with an intramuscular injection of a mix of xylazine and ketamine. The test item and negative control (PBS) were administered intravitreally in both eyes of anesthetized animals (3 μL per administration) on study day −3.
  • The positive control item (PBN) was injected intraperitoneally on Day 0, 4 times (0.5 h before starting light exposure, 2 h and 4 h after starting light exposure and just after the end of light exposure), at a dose volume of 2.5 mL/kg, using a 25-gauge needle mounted on a 1 mL-plastic syringe, protected from light.
  • Light Exposure
  • The rats were dark adapted for 36 hours and then exposed to a continuous blue fluorescent light (400-540 nm) in clear plastic cages for 6 hours. After exposure, the rats were placed in dark room for 24 hours before returning to standard cyclic light conditions.
  • Electroretinogram (ERG)
  • Electroretinograms (ERGs) were be recorded at baseline and on Day 14 on both eyes after overnight darkadaption. A-wave and b-wave amplitudes were measured for each ERG recording
  • Euthanasia
  • At the end of the in-life phase (Day 14), the animals were anesthetized and euthanized by intraperitoneal an overdose injection of pentobarbital.
  • Outer Nuclear Layer (ONL) Thickness Measurements
  • From the 10 main animals of each group, both eyeballs were enucleated, fixed in Bouin Hollande solution and embedded in paraffin. Thin sections (5 to 7 μm thick) were cut along the vertical meridian and stained with Trichrome-Masson. ONL thickness was measured at seven points (every 250 μm) from the optic nerve to the peripheral retina in each part (superior and inferior) of the retina. The thickness of the outer nuclear layer was measured at each point and the area under the curve (AUC) calculated.
  • Oligo Content Measurement and Quantification of Htra1 RNA Expression
  • Both eyeballs of 4 satellite animals from test article and PBS groups were used for bioanalysis. Immediately after euthanasia, Vitreous (V), Retina (R) and Choroid (CH) were quickly and carefully dissected out on ice and stored at −80° C. until shipment. Retina sample was lysed in 700 μL MagNa Pure 96 LC RNA Isolation Tissue buffer and homogenized by adding 1 stainless steel bead per 2 ml tube 2×1.5 min using a precellys evolution homogenizer followed by 30 min incubation at RT. The samples were centrifuges, 13000 rpm, 5 min. half was set aside for bioanalysis and for the other half, RNA extraction was continued directly.
  • For bioanalysis, the samples were diluted 10-50 fold for oligo content measurements with a hybridization ELISA method. A biotinylated LNA-capture probe and a digoxigenin-conjugated LNA-detection probe (both 35 nM in 5×SSCT, each complementary to one end of the LNA oligonucleotide to be detected) was mixed with the diluted homogenates or relevant standards, incubated for 30 minutes at RT and then added to a streptavidine-coated ELISA plates (Nunc cat. no. 436014).
  • The plates were incubated for 1 hour at RT, washed in 2×SSCT (300 mM sodium chloride, 30 mM sodium citrate and 0.05% v/v Tween-20, pH 7.0) The captured LNA duplexes were detected using an anti-DIG antibodies conjugated with alkaline phosphatase (Roche Applied Science cat. No. 11093274910) and an alkaline phosphatase substrate system (Blue Phos substrate, KPL product code 50-88-00). The amount of oligo complexes was measured as absorbance at 615 nm on a Biotek reader.
  • For RNA extraction, cellular RNA large volume kit (05467535001, Roche) was used in the MagNA Pure 96 system with the program: Tissue FF standard LV3.1 according to the instructions of the manufacturer, including DNAse treatment. RNA quality control and concentration were measured with an Eon reader (Biotek). The RNA concentration was normalized across samples, and subsequent cDNA synthesis and qPCR was performed in a one-step reaction using qScript XLT one-step RT-qPCR ToughMix Low ROX, 95134-100 (Quanta Biosciences). The following TaqMan primer assays were used in a duplex reaction: Htra1, Rn00581870_m1 and Rn00668987_m1 and housekeeping genes, HPRT, Rn01527840_m1 and Tbp, Rn01455646_m1, from Life Technologies. The qPCR analyses were run on a ViiA7 machine (Life Technologies). Rats/group: 5, n=10 eyes. Each eye was treated as an individual sample. The relative Htra1 mRNA expression level is shown as % of control (PBS).
  • Histology
  • Both eyeballs of the remaining 2 satellite animals from test article and PBS groups were removed and fixed in 10% neutral buffered formalin for 24 hours, trimmed and embedded in paraffin. ISH RNAscope was performed as described in example 10.
  • Example 12, Rat In Vivo Efficacy Kinetic Study, 3, 7 and 14 Days of Treatment, Intravitreal (IVT) Injection, Single Dose
  • Knockdown (KD) at mRNA level was observed in the retina for 2 selected HTRA1 LNA oligonucleotides targeting the “hotspot” in human HTRA1 pre-mRNA between position 33042-33064 (SEQ ID NO 147). This was observed both with qPCR and ISH readouts (see FIGS. 7A and /B and the following table).
  • qPCR ISH
    Compound Days of Residual mRNA Residual mRNA
    ID treatment level Stdev n level Stdev N
    143.1 3 59 31 12 11 14 4
    7 34 28 12 12 8 4
    14 39 35 12 3 5 4
    145.3 3 30 29 12 7 5 4
    7 35 32 12 4 3 4
    14 30 21 12 10 9 4
  • The variation of the knockdown is relatively large, see the standard deviations listed in the table. The variation seems to be at the administration level which can be seen when plotting a dose response curve for oligo content vs. residual HTRA1 mRNA level (see FIG. 7C).
  • Animals
  • All experiments were performed on albino Sprague-Dawley rats.
  • Compounds and Dosing Procedures
  • The animals were anesthetized in isofluran. The test item and negative control (PBS) were administered intravitreally in both eyes of anesthetized animals (3 μL per administration) on study day 1.
  • Euthanasia
  • At the end of the in-life phase (study day 4, 8 or 15) the rats were anesthetized and euthanized by decapitation.
  • Oligo Content Measurement and Quantification of Htra1 RNA Expression
  • Oligo content measurement and quantification of Htra1 mRNA expression was performed as described in Example 10.
  • The relative residual Htra1 mRNA expression level is shown as % of control (PBS).
  • Histology
  • Histology was performed as described in Example 10.
  • Example 13. Cynomolgus Monkey (Non-Human Primate, NHP) In Vivo Pharmacokinetics and Pharmacodynamics (PK/PD) Study, 21 Days of Treatment, Intravitreal (IVT) Injection, Single Dose
  • Knockdown was observed for 1 selected HTRA1 LNA oligonucleotide, 145.3, targeting the “hotspot” in human HTRA1 pre-mRNA between position 33042-33064 both at mRNA in the retina and at protein level in the retina and in the vitreous (see FIG. 8).
  • Animals
  • All experiments were performed on Cynomolgus monkeys (Macaca fascicularis).
  • Compounds and Dosing Procedures
  • Buprenorphine analgesia was administered prior to, and two days after test compound injection. The animals were anesthetized with an intramuscular injection of ketamine and xylazine. The test item and negative control (PBS) were administered intravitreally in both eyes of anesthetized animals (50 μL per administration) on study day 1 after local application of tetracaine anesthetic.
  • Euthanasia
  • At the end of the in-life phase (Day 22) all monkeys were euthanized by intraperitoneal an overdose injection of pentobarbital.
  • Oligo Content Measurement and Quantification of Htra1 RNA Expression by qPCR
  • Immediately after euthanasia, eye tissues were quickly and carefully dissected out on ice and stored at −80° C. until shipment. Retina sample was lysed in 700 μL MagNa Pure 96 LC RNA Isolation Tissue buffer and homogenized by adding 1 stainless steel bead per 2 ml tube 2×1.5 min using a precellys evolution homogenizer followed by 30 min incubation at RT. The samples were centrifuged, 13000 rpm, 5 min. Half was set aside for bioanalysis and for the other half, RNA extraction was continued directly.
  • For bioanalysis, the samples were diluted 10-50 fold for oligo content measurements with a hybridization ELISA method. A biotinylated LNA-capture probe and a digoxigenin-conjugated LNA-detection probe (both 35 nM in 5×SSCT, each complementary to one end of the LNA oligonucleotide to be detected) was mixed with the diluted homogenates or relevant standards, incubated for 30 minutes at RT and then added to a streptavidine-coated ELISA plates (Nunc cat. no. 436014).
  • The plates were incubated for 1 hour at RT, washed in 2×SSCT (300 mM sodium chloride, 30 mM sodium citrate and 0.05% v/v Tween-20, pH 7.0) The captured LNA duplexes were detected using an anti-DIG antibodies conjugated with alkaline phosphatase (Roche Applied Science cat. No. 11093274910) and an alkaline phosphatase substrate system (Blue Phos substrate, KPL product code 50-88-00). The amount of oligo complexes was measured as absorbance at 615 nm on a Biotek reader.
  • For RNA extraction, cellular RNA large volume kit (05467535001, Roche) was used in the MagNA Pure 96 system with the program: Tissue FF standard LV3.1 according to the instructions of the manufacturer, including DNAse treatment. RNA quality control and concentration were measured with an Eon reader (Biotek). The RNA concentration was normalized across samples, and subsequent cDNA synthesis and qPCR was performed in a one-step reaction using qScript XLT one-step RT-qPCR ToughMix Low ROX, 95134-100 (Quanta Biosciences). The following TaqMan primer assays were used in singplex reactions: Htra1, Mf01016150_, Mf01016152_m1 and Rh02799527_m1 and housekeeping genes, ARFGAP2, Mf01058488_g1 and Rh01058485_m1, and ARL1, Mf02795431_m1, from Life Technologies. The qPCR analyses were run on a ViiA7 machine (Life Technologies). Eyes/group: n=3 eyes. Each eye was treated as an individual sample. The relative Htra1 mRNA expression level is shown as % of control (PBS).
  • Histology
  • Eyeballs were removed and fixed in 10% neutral buffered formalin for 24 hours, trimmed and embedded in paraffin.
  • For ISH analysis, sections of formalin-fixed, paraffin-embedded retina tissue 4 μm thick were processed using the fully automated Ventana Dicovery ULTRA Staining Module (Procedure: mRNA Discovery Ultra Red 4.0-v0.00.0152) using the RNAscope 2.5 VS Probe-Mmu-HTRA1, REF 486979, Advanced Cell Diagnostics, Inc. Chromogen used is Fastred, Hematoxylin H counterstain.
  • HTRA1 Protein Quantification Using a Plate-Based Immunoprecipitation Mass Spectrometry (IP-MS) Approach
  • Sample Preparation, Retina
  • Retinas were homogenized in 4 volumes (w/v) of RIPA buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 0.25% deoxycholic acid, 1% NP-40, 1 mM EDTA, Millipore) with protease inhibitors (Complete EDTA-free, Roche) using a Precellys 24 (5500, 15 s, 2 cycles). Homogenates were centrifuged (13,000 rpm, 3 min) and the protein contents of the supernatants determined (Pierce BCA protein assay)
  • Sample Preparation, Vitreous
  • Vitreous humors (300 μl) were diluted with 5× RIPA buffer (final concentration: 50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 0.25% deoxycholic acid, 1% NP-40, 1 mM EDTA) with protease inhibitors (Complete EDTA-free, Roche) and homogenized using a Precellys 24 (5500, 15 s, 2 cycles). Homogenates were centrifuged (13,000 rpm, 3 min) and the protein contents of the supernatants determined (Pierce BCA protein assay)
  • Plate-Based HTRA1 Immunoprecipitation and Tryptic Digest
  • A 96 well plate (Nunc MaxiSorp) was coated with anti-HTRA1 mouse monoclonal antibody (R&D MAB2916, 500 ng/well in 50 μl PBS) and incubated overnight at 4° C. The plate was washed twice with PBS (200 μl) and blocked with 3% (w/v) BSA in PBS for 30 min at 20° C. followed by two PBS washes. Samples (75 μg retina, 100 μg vitreous in 50 μl PBS) were randomized and added to the plate followed by overnight incubation at 4° C. on a shaker (150 rpm). The plate was then washed twice with PBS and once with water. 10 mM DTT in 50 mM TEAB (30 μl) were then added to each well followed by incubation for 1 h at 20° C. to reduce cysteine sulfhydryls. 150 mM iodoacetamide in 50 mM TEAB (5 μl) were then added to each well followed by incubation for 30 min at 20° C. in the dark in order to block cysteine sulfhydryls. 10 μl Digestion solution were added to each well (final concentrations: 1.24 ng/μl trypsin, 20 fmol/μl BSA peptides, 26 fmol/μl isotope-labeled HTRA1 peptides, 1 fmol/μl iRT peptides, Biognosys) followed by incubation overnight at 20° C.
  • HTRA1 Peptide Quantification by Targeted Mass Spectrometry (Selected Reaction Monitoring, SRM)
  • Mass spectrometry analysis was performed on an Ultimate RSLCnano LC coupled to a TSQ Quantiva triple quadrupole mass spectrometer (Thermo Scientific). Samples (20 μL) were injected directly from the 96 well plate used for IP and loaded at 5 μL/min for 6 min onto a Acclaim Pepmap 100 trap column (100 μm×2 cm, C18, 5 μm, 100 Å, Thermo Scientific) in loading buffer (0.5% v/v formic acid, 2% v/v ACN). Peptides were then resolved on a PepMap Easy-SPRAY analytical column (75 μm×15 cm, 3 μm, 100 Å, Thermo Scientific) with integrated electrospray emitter heated to 40° C. using the following gradient at a flow rate of 250 nL/min: 6 min, 98% buffer A (2% ACN, 0.1% formic acid), 2% buffer B (ACN+0.1% formic acid); 36 min, 30% buffer B; 41 min, 60% buffer B; 43 min, 80% buffer B; 49 min, 80% buffer B; 50 min, 2% buffer B. The TSQ Quantiva was operated in SRM mode with the following parameters: cycle time, 1.5 s; spray voltage, 1800 V; collision gas pressure, 2 mTorr; Q1 and Q3 resolution, 0.7 FWHM; ion transfer tube temperature 300° C. SRM transitions were acquired for the HTRA1 peptide “LHRPPVIVLQR” and an isotope labelled (L-[U-13C, U-15N]R) synthetic version, which was used an internal standard. Data analysis was performed using Skyline version 3.6

Claims (1)

1. An antisense oligonucleotide of 10-30 nucleotides in length, wherein said antisense oligonucleotide targets a HTRA1 nucleic acid and comprises a contiguous nucleotide region of 10-22 nucleotides which are at least 90% such as 100% complementarity to SEQ ID NO 147:
SEQ ID NO 147: 5′ CCAACAACCAGGTAAATATTTG 3′
US16/730,779 2016-07-01 2019-12-30 Antisense oligonucleotides for modulating htra1 expression Abandoned US20200399641A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/730,779 US20200399641A1 (en) 2016-07-01 2019-12-30 Antisense oligonucleotides for modulating htra1 expression

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP16177508 2016-07-01
EP16177508.5 2016-07-01
EP17170129 2017-05-09
EP17170129.5 2017-05-09
US15/635,629 US10519450B2 (en) 2016-07-01 2017-06-28 Antisense oligonucleotides for modulating HTRA1 expression
US16/730,779 US20200399641A1 (en) 2016-07-01 2019-12-30 Antisense oligonucleotides for modulating htra1 expression

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/635,629 Division US10519450B2 (en) 2016-07-01 2017-06-28 Antisense oligonucleotides for modulating HTRA1 expression

Publications (1)

Publication Number Publication Date
US20200399641A1 true US20200399641A1 (en) 2020-12-24

Family

ID=59295175

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/635,629 Active US10519450B2 (en) 2016-07-01 2017-06-28 Antisense oligonucleotides for modulating HTRA1 expression
US16/730,779 Abandoned US20200399641A1 (en) 2016-07-01 2019-12-30 Antisense oligonucleotides for modulating htra1 expression

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/635,629 Active US10519450B2 (en) 2016-07-01 2017-06-28 Antisense oligonucleotides for modulating HTRA1 expression

Country Status (21)

Country Link
US (2) US10519450B2 (en)
EP (1) EP3478839A1 (en)
JP (3) JP7060525B2 (en)
KR (1) KR20190025970A (en)
CN (1) CN109415732B (en)
AU (1) AU2017289204A1 (en)
BR (1) BR112018077321A2 (en)
CA (1) CA3026855A1 (en)
CL (1) CL2018003698A1 (en)
CO (1) CO2018013240A2 (en)
CR (1) CR20180606A (en)
IL (1) IL263489A (en)
MA (1) MA45620A (en)
MX (1) MX2018015722A (en)
PE (1) PE20190381A1 (en)
PH (1) PH12018502707A1 (en)
RU (1) RU2019101298A (en)
SG (2) SG10202005885VA (en)
TW (1) TW201803990A (en)
WO (1) WO2018002105A1 (en)
ZA (1) ZA201808291B (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2017282651B2 (en) 2016-06-21 2021-08-12 Orion Ophthalmology LLC Heterocyclic prolinamide derivatives
EP3472151A4 (en) 2016-06-21 2020-03-04 Orion Ophthalmology LLC Carbocyclic prolinamide derivatives
CA3026855A1 (en) 2016-07-01 2018-01-04 F. Hoffmann-La Roche Ag Antisense oligonucleotides for modulating htra1 expression
AU2018386527A1 (en) 2017-12-22 2020-04-30 Roche Innovation Center Copenhagen A/S Gapmer oligonucleotides comprising a phosphorodithioate internucleoside linkage
KR20200104302A (en) 2017-12-22 2020-09-03 로슈 이노베이션 센터 코펜하겐 에이/에스 New thiophosphoramidite
EP4074724A1 (en) 2017-12-22 2022-10-19 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorodithioate internucleoside linkage
AU2019218987A1 (en) 2018-02-12 2020-07-23 Ionis Pharmaceuticals, Inc. Modified compounds and uses thereof
WO2020007772A1 (en) * 2018-07-02 2020-01-09 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting gbp-1
WO2020109343A1 (en) * 2018-11-29 2020-06-04 F. Hoffmann-La Roche Ag Combination therapy for treatment of macular degeneration
WO2020109344A1 (en) * 2018-11-29 2020-06-04 F. Hoffmann-La Roche Ag Occular administration device for antisense oligonucleotides

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3756313B2 (en) 1997-03-07 2006-03-15 武 今西 Novel bicyclonucleosides and oligonucleotide analogues
CN1273476C (en) 1997-09-12 2006-09-06 埃克西康有限公司 Bi-and tri-cyclic nucleoside, nucleotide and oligonucleotide analoguse
AU5290499A (en) 1998-08-03 2000-02-28 Novartis Ag Human htra serine protease
ES2234563T5 (en) 1999-02-12 2018-01-17 Daiichi Sankyo Company, Limited New nucleoside and oligonucleotide analogs
NZ514348A (en) 1999-05-04 2004-05-28 Exiqon As L-ribo-LNA analogues
US6617442B1 (en) 1999-09-30 2003-09-09 Isis Pharmaceuticals, Inc. Human Rnase H1 and oligonucleotide compositions thereof
EP1569661B1 (en) 2002-11-18 2009-09-09 Santaris Pharma A/S Antisense design
AU2006249925A1 (en) 2005-05-24 2006-11-30 Isis Pharmaceuticals, Inc. Modulation of LMW-PTPase expression
WO2007031091A2 (en) 2005-09-15 2007-03-22 Santaris Pharma A/S Rna antagonist compounds for the modulation of p21 ras expression
PL2314594T3 (en) 2006-01-27 2014-12-31 Isis Pharmaceuticals Inc 6-modified bicyclic nucleic acid analogs
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
DK2066684T3 (en) 2006-05-11 2012-10-22 Isis Pharmaceuticals Inc 5'-Modified Bicyclic Nucleic Acid Analogs
CA2658853A1 (en) 2006-07-26 2008-01-31 Yale University Diagnosis and treatment of age related macular degeneration
US20100166743A1 (en) 2006-10-06 2010-07-01 University Of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
EP2913341A1 (en) 2006-12-22 2015-09-02 University of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
ES2388590T3 (en) 2007-05-30 2012-10-16 Isis Pharmaceuticals, Inc. Analogs of bicyclic nucleic acids with N-substituted aminomethylene bridge.
ES2386492T3 (en) 2007-06-08 2012-08-21 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
TW200911290A (en) * 2007-07-02 2009-03-16 Alcon Res Ltd RNAI-mediated inhibition of HTRA1 for treatment of macular degeneration
ES2376507T5 (en) 2007-07-05 2015-08-31 Isis Pharmaceuticals, Inc. 6-disubstituted bicyclic nucleic acid analogs
US20120142608A1 (en) 2007-11-01 2012-06-07 University of Iowa Research Foundation Corporation Rca locus analysis to assess susceptibility to amd and mpgnii
US8546556B2 (en) 2007-11-21 2013-10-01 Isis Pharmaceuticals, Inc Carbocyclic alpha-L-bicyclic nucleic acid analogs
EP2356129B1 (en) 2008-09-24 2013-04-03 Isis Pharmaceuticals, Inc. Substituted alpha-l-bicyclic nucleosides
US9012421B2 (en) 2009-08-06 2015-04-21 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
WO2011156202A1 (en) 2010-06-08 2011-12-15 Isis Pharmaceuticals, Inc. Substituted 2 '-amino and 2 '-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
FR2965278B1 (en) 2010-09-23 2014-10-10 Univ Caen Basse Normandie PROCESS FOR IN VITRO OR EX VIVO PRODUCTION OF CHONDROCYTES AND USES THEREOF
CN104136451A (en) 2011-09-07 2014-11-05 玛瑞纳生物技术有限公司 Synthesis and uses of nucleic acid compounds with conformationally restricted monomers
WO2013154798A1 (en) 2012-04-09 2013-10-17 Isis Pharmaceuticals, Inc. Tricyclic nucleic acid analogs
US20150141320A1 (en) 2012-05-16 2015-05-21 Rana Therapeutics, Inc. Compositions and methods for modulating gene expression
MY173826A (en) 2012-11-15 2020-02-24 Roche Innovation Ct Copenhagen As Oligonucleotide conjugates
EP3368578B1 (en) 2015-10-30 2021-03-17 H. Hoffnabb-La Roche Ag Anti-htra1 antibodies and methods of use thereof
CA3026855A1 (en) 2016-07-01 2018-01-04 F. Hoffmann-La Roche Ag Antisense oligonucleotides for modulating htra1 expression
JP7033591B2 (en) 2016-11-11 2022-03-10 ロシュ イノベーション センター コペンハーゲン エーエス Capture and detection of therapeutic oligonucleotides
US20190055564A1 (en) 2017-06-01 2019-02-21 F. Hoffmann-La Roche Ag Antisense oligonucleotides for modulating htra1 expression

Also Published As

Publication number Publication date
EP3478839A1 (en) 2019-05-08
CN109415732A (en) 2019-03-01
MX2018015722A (en) 2019-05-27
CA3026855A1 (en) 2018-01-04
RU2019101298A (en) 2020-08-03
ZA201808291B (en) 2019-10-30
SG10202005885VA (en) 2020-07-29
JP2022106785A (en) 2022-07-20
CR20180606A (en) 2019-02-14
JP7060525B2 (en) 2022-04-26
JP2024029178A (en) 2024-03-05
CL2018003698A1 (en) 2019-04-05
KR20190025970A (en) 2019-03-12
US20180002701A1 (en) 2018-01-04
RU2019101298A3 (en) 2021-01-26
PE20190381A1 (en) 2019-03-08
US10519450B2 (en) 2019-12-31
JP2019522987A (en) 2019-08-22
AU2017289204A1 (en) 2019-01-17
JP7416852B2 (en) 2024-01-17
IL263489A (en) 2019-01-31
CN109415732B (en) 2024-01-02
PH12018502707A1 (en) 2019-11-11
CO2018013240A2 (en) 2018-12-14
BR112018077321A2 (en) 2019-04-24
SG11201811554SA (en) 2019-01-30
WO2018002105A1 (en) 2018-01-04
MA45620A (en) 2019-05-08
TW201803990A (en) 2018-02-01

Similar Documents

Publication Publication Date Title
US10519450B2 (en) Antisense oligonucleotides for modulating HTRA1 expression
US11542501B2 (en) Antisense oligonucleotides targeting ATXN3
US20220042022A1 (en) Antisense oligonucleotides for modulating htra1 expression
US11066669B2 (en) Oligonucleotides for modulating ATXN2 expression
US20210123054A1 (en) Oligonucleotides for modulating tau expression
US20210238608A1 (en) Oligonucleotides for modulating scn9a expression
US20190365795A1 (en) Oligonucleotides for modulating atxn2 expression
US11485975B2 (en) Oligonucleotides for modulating TMEM106B expression
WO2020201339A1 (en) Oligonucleotides for modulating atxn2 expression
US20200378970A1 (en) Companion diagnostic for htra1 rna antagonists
US11286485B2 (en) Oligonucleotides for modulating ATXN2 expression
NZ749395A (en) Antisense oligonucleotides for modulating htra1 expression
US20220177883A1 (en) Antisense Oligonucleotides Targeting ATXN3
US20210261961A1 (en) Antisense oligonucleotides targeting tia1

Legal Events

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

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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