WO2009039442A1 - Nfia in glial fate determination, glioma therapy and astrocytoma treatment - Google Patents

Nfia in glial fate determination, glioma therapy and astrocytoma treatment Download PDF

Info

Publication number
WO2009039442A1
WO2009039442A1 PCT/US2008/077119 US2008077119W WO2009039442A1 WO 2009039442 A1 WO2009039442 A1 WO 2009039442A1 US 2008077119 W US2008077119 W US 2008077119W WO 2009039442 A1 WO2009039442 A1 WO 2009039442A1
Authority
WO
WIPO (PCT)
Prior art keywords
nfia
inhibitor
glioma
antisense
shrna
Prior art date
Application number
PCT/US2008/077119
Other languages
French (fr)
Other versions
WO2009039442A9 (en
Inventor
David J. Anderson
Ben Deneen
Hae-Ri Song
Original Assignee
California Institute Of Technology
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 California Institute Of Technology filed Critical California Institute Of Technology
Publication of WO2009039442A1 publication Critical patent/WO2009039442A1/en
Publication of WO2009039442A9 publication Critical patent/WO2009039442A9/en

Links

Classifications

    • 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
    • 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
    • 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
    • C12N2310/111Antisense spanning the whole gene, or a large part of it
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed

Definitions

  • Embodiments disclosed herein relate to molecular medicine, and in particular to compositions and methods that can be used for glioma and astrocytoma therapy and treatment. Description of the Related Art
  • Gliomas tumors derived from glia cells of the central nervous system (CNS) account for approximately 30% of all primary brain tumors in adults. The median survival rate is about 12 months. Gliomas are usually treated by surgery, chemotherapy, radiotherapy and/or fractionated stereotype radio surgery.
  • Gliomas are heterogeneous in their cellular content and can be divided into groups of astrocytomas, anablastic astrocytomas and glioblastoma multiformes.
  • Traditional approaches to therapeutic intervention have relied upon either surgery, chemotherapy, or radiotherapy.
  • Recent advances in molecular genetics have revealed many genetic mutations and associated signaling pathways that may play a causative role in the generation of gliomas. While these advances have provided numerous candidate pathways that can be utilized in the development of rational therapy rooted in the biology of the disease, it remains to be seen whether such approaches will come to fruition. As such, there is a continued need for gliomal therapies.
  • NFIA Nuclear Factor I family of genes are glial fate determinants in the developing central nervous system. Interestingly, NFIA is also expressed in all grades of human astroctyoma, the most deadly form of glioma. Described herein is Applicants' discovery that NFIA plays a causative role in glioma fo ⁇ nation and Applicants' discovery of therapeutics based on the elucidation of NFIA' s role in glioma formation.
  • glioma therapy and treatment relate to methods for reducing the size of a glioma. Some embodiments relate to methods of arresting or slowing the development or progression of gliomal tumors. In some embodiments, the gliomas can be astrocytomas.
  • the methods described herein include the steps of contacting a glioma with a therapeutically effective amount of an NFlA inhibitor.
  • the NFIA inhibitor can include an NFIA antisense polynucleotide, such as an NFIA short hairpin RNA (shRNA).
  • the shRNA NFlA inhibitor can target human NFIA.
  • the NFIA inhibitor can be an NFIA shRNA that comprises the sequence of SEQ ID NO: 1.
  • the contacting step comprises contacting the glioma with a viral expression vector that encodes an NFIA antisense polynucleotide, such as a lentiviral expression vector.
  • the NFIA inhibitor can be an antibody that specifically binds to NFIA.
  • NFIA antisense polynucleotide comprises an isolated polynucleotide comprising the sequence of SEQ ID NO: 1. BRIEF DESCRIPTION OF THE DRAWINGS
  • Figure IA is a bar graph showing the relative expression levels of NFIA in U87 glioma cells that were either untreated, treated with a mutant NFIA shRNA, or treated with an NFIA shRNA that targets human NFIA.
  • Figure 2 is a graph showing the growth (as expressed in total cell number) of U87 glioma cells infected with either a viral construct encoding a human NFIA shRNA or a mutant version of the human NFIA shRNA, or that were not infected.
  • Figure 3 is a graph showing the size and frequency of tumors in mice receiving transplants of: U87 glioma cells transduced with a lentiviral expression vector encoding NFIA cDNA; untreated U87 cells, U87 cells transduced with either a lentiviral construct encoding a NFIA shRNA; or U87 cells transduced with a lentiviral construct encoding a mutant version of the NFIA shRNA.
  • Figures 4A-4D are images of MRIs of genetically modified U87 cells implanted into a mouse brain, 28 days post implantation.
  • the arrows denote tumor growth.
  • the embodiments disclosed herein relate to the treatment of gliomas and are based, in part, on Applicants' discovery that Nuclear Factor IA (NFIA) plays a role in glioma formation.
  • NFIA Nuclear Factor IA
  • Embodiments herein relate to methods for treating gliomas, for reducing the tumor size of gliomas, and for reducing the functional NFIA levels in gliomas with NFlA inhibitors.
  • NFIA inhibitor refers to a compound or composition that reduces the functional NFlA levels in a target cell (e.g., a glioma cell).
  • NF ⁇ A inhibitors can function by inhibiting the expression of NFIA, for example by inhibiting transcription, translation, or processing of NFIA, or by inhibiting a functional activity of NFIA, such as DNA binding.
  • NFIA inhibitors include, but are not limited to, antisense polynucleotides, antibodies, and small molecule inhibitors of NFIA.
  • NFIA Antisense Polynucleotides include, but are not limited to, antisense polynucleotides, antibodies, and small molecule inhibitors of NFIA.
  • an NFIA inhibitor can be an NFIA antisense polynucleotide.
  • antisense polynucleotides useful in the embodiments disclosed herein include single-stranded DNAs and RNAs that bind to complementary target mRNA and inhibit translation and/or induce RNaseH-mediated degradation of the target transcript; siRNA oligonucleotides, which are short, double-stranded RNAs that activate the RNA interference (RNAi) pathway leading to target mRNA degradation; ribozymes, which are oligonucleotide-based endonucleases that are designed to cleave specific mRNA transcripts; and nucleic acid aptainers and decoys, which are non-naturally occurring oligonucleotides that bind to and block protein targets in a manner analogous to small molecule drugs.
  • RNAi RNA interference
  • oligonucleotide refers to an oligomer or polymer of nucleic acids such as ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or mimetic s thereof.
  • RNA ribonucleic acid
  • DNA deoxyribonucleic acid
  • oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent intemucleoside (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions which function similarly. Modified or substituted oligonucleotides provide desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target and increased stability in the presence of nucleases.
  • antisense oligonucleotides that comprise only naturally occurring nucleobases
  • the antisense oligonucleotides provided herein can be oligomeric oligonucleotide mimetics.
  • antisense compounds provided herein include oligonucleotides containing modified backbones or non- natural internucleoside linkages. Oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone.
  • the antisense polynucleotides comprise oligomeric oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotri esters, aminoalkylphosphotri-esters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates, 5'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriest- ers, selenophosphates and borano- phosphates having normal 3'-5' linkages, T-S" linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a
  • Oligonucleotides having inverted polarity can comprise a single 3' to 3' linkage at the 3'-most internucleotide linkage i,e. a single inverted nucleoside residue which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof).
  • Various salts, mixed salts and free acid forms are also included.
  • NFIA antisense polynucleotides can comprise modified oligonucleotide backbones that do not include a phosphorus atom.
  • NFIA antisense polynucleotides can have backbones formed by short chain alkyl or cycloalkyl intemucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
  • morpholino linkages formed in part from the sugar portion of a nucleoside
  • siloxane backbones sulfide, sulfoxide and sulfone backbones
  • formacetyl and thioformacetyl backbones methylene formacetyl and thioformacetyl backbones
  • riboacetyl backbones alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH 2 component parts.
  • Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos.: 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; 5,792,608; 5,646,269 and 5,677,439, certain of which are commonly owned with this application, and each of which is herein incorporated by reference.
  • NFIA antisense polynucleotides can comprise mimetics wherein both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with non-naturally occurring groups.
  • the base units are maintained for hybridization with an appropriate target polynucleotides.
  • some embodiments provide NFlA antisense polynucleotides that comprise a peptide nucleic acid (PNA).
  • PNA peptide nucleic acid
  • the sugar-backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone.
  • nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
  • Representative United States patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos.: 5,539,082; 5,714,331 ; and 5,719,262, each of which is herein incorporated by reference in its entirety. Further teaching of PNA compounds can be found in Nielsen et al., Science, 1991, 254, 1497-1500.
  • NFIA antisense oligonucleotides can have phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular -CH 2 -NH-O-CH 2 -, -CH 2 -N(CHs)-O-CH 2 - [known as a methylene (methylimino) or MMI backbone], -CH 2 -O-N(CH 3 )-CH 2 -, -CH 2 -N(C H3 J-N(CH 3 )- CH 2 -- and — O— N(CH 3 )- CH 2 - CH 2 - [wherein the native phosphodi ester backbone is represented as -0-P-O-CH 2 -] of the above referenced U.S.
  • the NFIA antisense polynucleotides can contain one or more substituted sugar moieties.
  • the antisense oligonucleotides disclosed herein comprise one of the following at the T position: OH; F; O— , S-, or N-alkyl; 0—, S-, or N-alkenyl; O— , S— or N-alkynyl; or O-alkyl-0-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C] to C
  • the antisense oligonucleotides can include one of the following groups at the T position: O[(CH 2 ) n O] m CH 3 , O(CH 2 ) n OCH 3 , O(CH 2 ) n NH 2 , O(CH 2 ) n CH 3 , O(CH 2 ) n ONH 2 , and 0(CH 2 ) n ON[(CH 2 ) n CH 3 )] 2 , where n and m are from 1 to about 10.
  • the antisense oligonucleotides comprise one of the following at the 2' position: Ci to C 1O lower alkyl, substituted lower alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH 5 SCH 3 , OCN, Cl, Br, CN, CF 3 , OCF 3 , SOCH 3 , SO 2 CH 3 , ONO 2 , NO 2 , N3, NH 2 , heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties.
  • the antisense oligonucleotides include T- methoxyethoxy (2'-0-CH 2 CH 2 OCH 3 , also known as 2'-O-(2-methoxyethyl) or 2'-MOE) (Martin et al., HeIv. Chim. Acta, 1995, 78, 486-504) i.e., an alkoxyalkoxy group.
  • the antisense polynucleotides or oligonucleotides disclosed herein include T- dim ethyl aminooxyethoxy, i.e., a O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2'-DMAOE, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-O-dimethylaminoethoxyethyl or 2'-DMAEOE), i.e., 2'-O ⁇ CH 2 -O-CH 2 -N(CH 2 ) 2 , also described in examples herein below.
  • T- dim ethyl aminooxyethoxy i.e., a O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2'-DMAOE
  • 2'-dimethylaminoethoxyethoxy also known in the art as 2'-O-dimethylaminoethoxyethyl or 2'-DMAE
  • the NFIA antisense polynucleotides and oligonucleotides can include Locked Nucleic Acids (LNAs) in which the 2'-hydroxyl group is linked to the 3' or 4' carbon atom of the sugar ring thereby forming a bicyclic sugar moiety.
  • the linkage can be a methelyne (-CH 2 - ) n group bridging the T oxygen atom and the 4' carbon atom wherein n is 1 or 2.
  • LNAs and preparation thereof are described in WO 98/39352 and WO 99/14226, each of which is herein incorporated by reference in its entirety.
  • the NFIA antisense polynucleotides and oligonucleotides can include 2'-methoxy (2'-0-CH 3 ), 2'-aminopropoxy (T- OCH 2 CH 2 CH 2 NH 2 ), 2'-a!lyl (2'-CH 2 -CH.dbd.CH 2 ), 2'-O-aHyl (2'-O-CH 2 -CH.dbd.CH 2 - ) and 2'-fluoro (2'-F).
  • the 2 '-modification may be in the arabino (up) position or ribo (down) position.
  • the 2'-arabino modification is 2'-F.
  • oligonucleotide Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'- 5 linked oligonucleotides and the 5' position of 5' terminal nucleotide. Oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative United States patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat.
  • Oligonucleotides may also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions.
  • nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U).
  • Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-amino adenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouraciI, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyI and other 8-substituted adenines and guanines, 5-halo particularly 5-bro
  • nucleobases include tricyclic pyrimidines such as phenoxazine cytidine(lH-pyrimido[5,4-b][l,4]benzoxazi- n-2(3H)-one), phenothiazine cytidine (lH-pyrimido[5,4-b][l,4]benzothiazin- 2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g.
  • nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Further nucleobases include those disclosed in U.S.
  • 5-substituted pyrimidines include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2- aminopropyladenine, 5-propynyluracil and 5-propynylcytosine.
  • 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2 0 C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., eds., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are presently preferred base substitutions, even more particularly when combined with 2'-O-methoxyethyl sugar modifications.
  • the NFIA antisense oligonucleotides can be chemically linked to one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide.
  • the NFlA antisense oligonucleotides can include conjugate groups covalently bound to functional groups such as primary or secondary hydroxyl groups.
  • conjugate groups include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers, and groups that reduce the anionic charge of the polynucleotides, thereby enhancing transport across cellular membranes.
  • Conjugate groups include cholesterols, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes.
  • Groups that enhance the pharmacodynamic properties include groups that improve oligomer uptake, enhance oligomer resistance to degradation, and/or strengthen sequence-specific hybridization with RNA.
  • Groups that enhance the pharmacokinetic properties include groups that improve oligomer uptake, distribution, metabolism or excretion. Representative conjugate groups are disclosed for example in International Patent Application PCTYUS92/09196, filed Oct. 23, 1992, and PCT/US2007/003641, filed Feb. 9, 2007, the entire disclosures of which are incorporated herein by reference in their entireties.
  • lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Let., 1994, 4, 1053-1060), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660, 306-309; Manoharan et al., Bioorg. Med. Chem.
  • lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Let., 1994, 4, 1053-1060), a thioether, e
  • Acids Res., 1990, 18, 3777-3783 a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969- 973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp.
  • Oligonucleotides of the invention may also be conjugated to active drug substances, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fenbufen, ketoprofen, (S)-(+)-pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indomethicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic, for example.
  • active drug substances for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fenbufen, ketoprofen, (S)-(+)-pranoprofen, car
  • Oligonucleotide-drug conjugates and their preparation are described for example in U.S. patent application Ser. No. 09/334,130 (filed Jun. 15, 1999) which is incorporated herein by reference in its entirety. [0034] Representative United States patents that teach the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Pat.
  • NFIA antisense oligonucleotide or polynucleotide can be modified.
  • the NFlA antisense oligonucleotides do not contain any modified units.
  • the NFIA antisense oligonucleotides disclosed herein are not uniformly modified. In some embodiments, more than one of the aforementioned modifications can be incorporated in a single NFIA antisense polynucleotide or oligonucleotide, or even at a single nucleoside within an oligonucleotide.
  • the antisense compounds are chimeric.
  • the term “chimeric” antisense compounds or “chimeras,” refer to antisense compounds, particularly oligonucleotides, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an oligonucleotide compound. These oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid.
  • An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids.
  • RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene expression. Consequently, comparable results can often be obtained with shorter oligonucleotides when chimeric oligonucleotides are used, compared to phosphorothioate deoxyoligonucleotides hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
  • chimeric NFIA antisense compounds can be formed as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above. Such compounds have also been referred to in the art as hybrids or gapmers. Representative United States patents that teach the preparation of such hybrid structures include, but are not limited to, U.S. Pat.
  • NFIA antisense compounds disclosed herein can be conveniently and routinely made through the well-known technique of solid phase synthesis.
  • Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, Calif.), BioAutomation (Irving, TX), or the like. Any other means for such synthesis known in the art may additionally or alternatively be employed. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and alkylated derivatives.
  • NFIA antisense polynucleotides and oligonucleotides provided herein include any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof.
  • NFlA antisense polynucleotides and oligonucleotides can include prodrugs and pharmaceutically acceptable salts of the antisense polynucleotides, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
  • prodrug indicates a therapeutic agent that is prepared in an inactive form that is converted to an active form within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or conditions.
  • some embodiments provide prodrug versions of the NFlA antisense polynucleotides and oligonucleotides prepared as SATE [(S-acetyl-2-thioethyl) phosphate] derivatives according to the methods disclosed in WO 93/24510 to Gosselin et al., published Dec. 9, 1993 or in WO 94/26764 and U.S. Pat. No. 5,770,713 to Imbach et al.
  • the NFIA antisense polynucleotides can be 5-10- mers, 10-15-mers, 15-20mers, 20-25 mers, 25-30-mers, 30-40-mers, 40-45-mers, 45-50-mers, 50-55-mers, 55-60-mers, 60-65-mers, 65-70-mers, 70-75-mers, 75-80-mers, 80-85-mers, 85- 90-mers, 90-95-mers, 95-100-mers, 100-120-mers, 120-140-mers, 140-160-mers, 160-180- mers, 180-200-mers, or greater, or any number in between, including full length genes or RNA transcripts thereof, hi some embodiments the NFIA antisense polynucleotide can be, for example, a 43-55-mer that forms a hairpin, wherein about 4-5 of the nucleotide/modified subunits form the turn.
  • NFIA antisense polynucleotides target human NFlA.
  • human NFlA Several sequences of human NFlA are publicly available, and can be found, for example, under GENBANKTM accession numbers NM 005595 (SEQ ID NO: 3); NM 001 134673 (SEQ ID NO: 4).
  • NFIA antisense polynucleotides target NFIA from a different organism, such as a different mammal, such of mice, rats, rabbits, guinea pigs, dogs, cats, sheep, goats, cows, primates, such as monkeys, chimpanzees, and apes, and the like.
  • antisense NFlA polynucleotides can be designed to target any NFIA sequence now known or discovered in the future
  • NFIA antisense polynucleotides that comprise NFIA shRNA sequences.
  • Exemplary NFIA shRNA antisense polynucleotides include, but are not limited to, polynucleotides that comprise, consist essentially of, or consist of at least about 20 consecutive nucleic acids of the nucleic acid sequence of SEQ ID NO: 1, or mimetics thereof.
  • the NFIA shRNA can have at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity to at least 20 nucleotides of the nucleic acid sequence of SEQ ID NO: 1. Sequence identity can be determined using mathematical algorithms known to those skilled in the art. A non-limiting example of a mathematical algorithm utilized for comparison of sequences is the algorithm of Karlin S and Altschul S F, Proc. Natl. Acad. Sci. USA 87:2264-68 (1990), modified as in Karlin S and Altschul S F, Proc. Natl.
  • NFIA siRNA antisense targets are generally 15-25 bases long, and refer to the region of an mRNA to which an antisense compound hybridizes. Determination of regions of the NFIA coding sequence useful as NFIA inhibitors is NFIA siRNA antisense targets can be selected using routine methods known to those skilled in the art. For example, siRNA targets are routinely identified by scanning an mRNA sequence for AA dinucleotides and recording the 19 nucleotides immediately downstream of the AA. Other methods can also been used to select the siRNA targets.
  • the selection of the siRNA target sequence is purely empirically determined (see, e.g., Sui G et al., Proc. Natl. Acad. Sci. USA 99:5515-20 (2002)), as long as the target sequence starts with GG and does not share significant sequence homology with other genes as analyzed by BLAST search.
  • a more elaborate method is employed to select the siRNA target sequences. This procedure exploits an observation that any accessible site in endogenous mRNA can be targeted for degradation by synthetic oligodeoxyribonucleotide/RNase H method (see, e.g., Lee N S et al., Nature Biotechnol. 20:500-05 (2002)).
  • an NFIA antisense hairpin siRNA expression cassette is constructed to contain the sequence corresponding to the target, followed by a short spacer, the antisense strand of the target, and 5-6 Ts as transcription terminator.
  • the order of the sense and antisense strands within the siRNA expression constructs can be altered without affecting the gene silencing activities of the hairpin siRNA. Lm certain instances, the reversal of the order may cause partial reduction in gene silencing activities.
  • the length of nucleotide sequences that serve as each side of the stem of the hairpin siRNA expression cassette can range, for instance, from 19 to 29 or more.
  • the loop size can range from 3 to 23 nucleotides.
  • the stem of the hairpin can be greater than 29 bases long, e.g., 30-50, bases, or more.
  • the stem of the hairpin can be less than 29 bases, e.g. 5-28 bases.
  • the loop can be greater than 23 bases long, e.g., 24-50 bases.
  • a 5' overhang in the hairpin siRNA construct can be used, provided that the hairpin siRNA provides the descired therapeutic benefit, or reduces the levels of functional NFIA as described herein.
  • the 5' overhang includes about 6 nucleotide residues.
  • the target sequence for RNAi is a 21-mer sequence.
  • the 5' end of the target sequence has the dinucleotide "NA", where "N” can be any base and "A" represents adenine.
  • the remaining 19-mer sequence has a GC content of between 35% and 55%.
  • the remaining 19-mer sequence does not include any four consecutive A or T (i.e., AAAA or TTTT), three consecutive G or C (i.e., GGG or CCC), or seven "GC” in a row.
  • Additional criteria can also be used for selecting RNAi target sequences. For instance, the GC content of the remaining 19-mer sequence can be limited to between 45% and 55%.
  • any 19-mer sequence having three consecutive identical bases i.e., GGG, CCC, TTT, or AAA
  • the remaining 19-mer sequence can be selected to have low sequence homology to other genes.
  • potential target sequences are searched by BLASTN against NCBFs human UniGene cluster sequence database.
  • the human UniGene database contains non-redundant sets of gene-oriented clusters. Each UniGene cluster includes sequences that represent a unique gene. 19-mer sequences producing no hit to other human genes under the BLASTN search can be selected.
  • an NFIA siRNA sequence of the present invention can be introduced into a cell that expresses the NF3A gene.
  • the polypeptide or mRNA level of the NFIA gene in the cell can be detected.
  • a substantial change in the expression level of the NFIA gene before and after the introduction of the siRNA sequence is indicative of the effectiveness of the siRNA sequence in suppressing the expression of the NFlA gene.
  • the expression levels of other genes are also monitored before and after the introduction of the siRNA sequence.
  • siRNA sequence which has inhibitory effect on NFlA gene expression but does not significantly affect the expression of other genes can be selected.
  • multiple siRNA or other RNAi sequences can be introduced into the same target cell. These siRNA or RNAi sequences specifically inhibit NFIA gene expression but not the expression of other genes.
  • siRNA or other RNAi sequences that inhibit the expression of the NFIA gene and one or more additional genes can be used.
  • an NFIA inhibitor can be an anti-NFIA antibody.
  • antibody is used in the broadest sense and specifically covers, for example, single anti-NFIA monoclonal antibodies (e.g. antagonist, and neutralizing antibodies), anti-NFIA antibody compositions with polyepitopic specificity, single chain anti-NFIA antibodies, and fragments of anti-NFIA antibodies (see below).
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts.
  • NFIA antibodies useful in the embodiments described herein are commercially available, or can be generated using routine methods known to those skilled in the art.
  • Routine screening can be used to identify anti-NFlA-antibodies that are useful in the methods described herein, e.g., to reduce the activity of NFIA, to reduce tumor size, to treat gliomas, etc. Such screening methods include those detailed in the examples herein.
  • Polyclonal anti-NFIA antibodies can be prepared as described above by immunizing a suitable subject with an NFIA immunogen.
  • the anti-NFIA antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELlSA) using immobilized NFIA polypeptide.
  • ELlSA enzyme linked immunosorbent assay
  • the antibody molecules directed against a NFIA polypeptide can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as protein A chromatography to obtain the IgG fraction.
  • antibody- producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler G and Milstein C, Nature 256:495-97 (1975) (see also, Brown J P et al., J. Immunol. 127:539- 46 (1981): Brown J P et al., J. Biol. Chem. 255:4980-83 (1980); Yeh M Y et al., Proc. Natl. Acad. Sci. USA 76:2927-31 (1979); Yeh M Y et al., Int. J.
  • an immortal cell line typically a myeloma
  • lymphocytes typically splenocytes
  • the culture supernatants of the resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds specifically to a Ccdc80 polypeptide
  • any of the many well known protocols used for fusing lymphocytes and immortalized cell lines can be applied for the purpose of generating an anti-NFIA monoclonal antibody (see, e.g., Galfre G et al., Nature 266:550-52 (1977); Geifer M L et ah, supra; Lerner E A, supra; Kenneth, Monoclonal Antibodies, supra).
  • the immortal cell line e.g., a myeloma cell line
  • the immortal cell line is derived from the same mammalian species as the lymphocytes.
  • murine hybridomas can be made by fusing lymphocytes from a mouse immunized with an immunogenic preparation of the present invention with an immortalized mouse cell line.
  • Preferred immortal cell lines are mouse myeloma cell lines that are sensitive to culture medium containing hypoxanthine, aminopterin and thymidine ("HAT medium"). Any of a number of myeloma cell lines may be used as a fusion partner according to standard techniques, for example, the P3-NSl/l-Ag4-l , P3-x63-Ag8.653 or Sp2/0-Agl4 myeloma lines. These myeloma lines are available from the American Type Culture Collection (ATCC), Rockville, MD.
  • ATCC American Type Culture Collection
  • HAT-sensitive mouse myeloma cells are fused to mouse splenocytes using polyethylene glycol ("PEG").
  • PEG polyethylene glycol
  • Hybridoma cells resulting from the fusion are then selected using HAT medium, which kills unfused and unproductively fused myeloma cells (unfused splenocytes die after several days because they are not transformed).
  • Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind a NFLA molecule, for example, using a standard ELISA assay.
  • a monoclonal anti-NFIA antibody can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with NFIA to thereby isolate immunoglobulin library members that bind a NFIA polypeptide.
  • Kits for generating and screening phage display libraries are commercially available (e.g., the GE Healthcare Recombinant Phage Antibody System, Catalog No. 27-9400-01).
  • examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S. Pat. No.
  • recombinant anti-NFIA antibodies such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions
  • recombinant DNA techniques known in the art, for example using methods described in WO 87/02671 ; EP 0 184 187; EP 0 171 496; EP 0 173 494; WO 86/01533; U.S. Pat. No. 4,816,567; EP 0 125 023; Better M et al., Science 240: 1041-43 (1988); Liu A Y et al., Proc. Natl. Acad. Sci.
  • humanized antibodies can be made according to standard protocols such as those disclosed in U.S. Pat. No, 5,565,332.
  • antibody chains or specific binding pair members can be produced by recombination between vectors comprising nucleic acid molecules encoding a fusion of a polypeptide chain of a specific binding pair member and a component of a replicable genetic display package and vectors containing nucleic acid molecules encoding a second polypeptide chain of a single binding pair member using techniques known in the art, for example, as described in U.S. Pat. No. 5,565,332; 5,871,907; or 5,733,743.
  • anti-NFIA antibodies that recognize intracellular NFIA can be used, e.g., intracellular ⁇ to inhibit NFIA protein activity.
  • intracellular antibodies to inhibit protein function in a cell is known in the art (see e.g., Carlson J R 3 MoI. Cell. Biol. 8:2638-46 (1988); Biocca S et al., EMBO J. 9:101-08 (1990); Werge T M et al., FEBS Lett. 274: 193-98 (1990); Carlson J R, Proc. Natl. Acad. Sci. USA 90:7427-28 (1993); Marasco W A et al., Proc. Natl. Acad. Sci.
  • a recombinant expression vector is prepared which encodes the antibody chains in a form such that, upon introduction of the vector into a cell, the antibody chains are expressed by the cell as a functional antibody.
  • an intracellular antibody that specifically binds NFIA protein is preferably secreted from the cell.
  • antibody light and heavy chain cDNAs encoding antibody chains specific for the target protein of interest are isolated, typically from a hybridoma that secretes a monoclonal antibody specific for the NFIA protein.
  • Hybridomas secreting anti-NFIA monoclonal antibodies, or recombinant anti-NFIA monoclonal antibodies can be prepared as described above.
  • DNAs encoding the light and heavy chains of the monoclonal antibody are isolated by standard molecular biology techniques.
  • hybridoma derived antibodies light and heavy chain cDNAs can be obtained, for example, by PCR amplification or cDNA library screening.
  • cDNA encoding the light and heavy chains can be recovered from the display package (e.g., phage) isolated during the library screening process.
  • Nucleotide sequences of antibody light and heavy chain genes from which PCR primers or cDNA library probes can be prepared are known in the art. For example, many such sequences are disclosed in Kabat E A et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242 and in the "Vbase" human germline sequence database.
  • the antibody light and heavy chain sequences are cloned into a recombinant expression vector using standard methods.
  • An antibody expression vector can encode an antibody in one of several different forms. For example, in one embodiment, the vector encodes full-length antibody light and heavy chains such that a full-length antibody is expressed.
  • the expression vector encoding the anti-NFIA intracellular or extracellular antibody is introduced into the cell by standard transfection methods, as discussed herein.
  • Anti-NFIA antibodies that function as NFIA inhibitors can be identified by screening for a desired inhibitory activity, for example, by using the methods described in the examples and elsewhere in the disclosure.
  • anti-NFIA antibodies can be screened in vitro for their ability to slow or arrest growth of gliomas, to decrease NFIA expression in gliomal cells, inhibiot NFIA activity (e.g., inhibit transcription of NFIA- induced genes, etc),or the like.
  • the methods disclosed herein involve contacting a glioma with an effective amount of an NFlA inhibitor.
  • effective amount refers to the amount of an NFIA inhibitor that is effective to at least slow the rate of glioma tumor growth, slow or arrest the progression of glioma, or decrease glioma tumor size. Tumor growth and tumor size can be measured using routine methods known to those skilled in the art, including, for example, magnetic resonance imagine and the like.
  • Gliomas can be subdivided based on the WHO grading system into grade HV 3 with grade I reflecting a benign, highly differentiated tumor and grades H-IV marking increasing states of malignancy, and decreasing degrees of differentiation.
  • an "effective amount" of an NFIA inhibitor is an amount effective to result in a downgrading of a tumor, or an amount effective to slow or prevent the progression of a glioma to a higher grade.
  • an “effective amount” can refer to the amount of NFIA inhibitor necessary to cause "knock-down" of NFIA expression relative to levels present following administration or expression of a non-targeting control RNA (e.g., a non-targeting control siRNA). Knockdown of expression of an amount including and between 50% and 100%, e.g., 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or more, is contemplated by embodiments herein.
  • Knock-down can be assessed by measuring the mRNA levels using quantitative polymerase chain reaction (qPCR) amplification or by measuring protein levels by western blot or enzyme-linked immunosorbent assay (ELISA). Analyzing the protein level provides an assessment of both mRNA cleavage as well as translation inhibition. Further techniques for measuring knock-down include RNA solution hybridization, nuclease protection, northern hybridization, gene expression monitoring with a microarray, antibody binding, radioimmunoassay, and fluorescence activated cell analysis.
  • qPCR quantitative polymerase chain reaction
  • ELISA enzyme-linked immunosorbent assay
  • the methods disclosed herein include contacting a glioma or glioma target cell with an effective amount of an NFIA inhibitor.
  • the NFIA inhibitor comprises a polynucleotide, (including recombinant expression vectors encoding NFIA antisense RNA, intracellular NFIA antibodies, or dominant negative NFIA inhibitors)
  • the agents can be introduced into cells of the glioma using methods known in the art for introducing polynucleotides (e.g., DNA, RNA or the like) into cells in vivo. Examples of such methods encompass both non-viral and viral methods, including but not limited to the methods described below.
  • Naked polynucleotides can be introduced into cells or glioma in vivo by directly injecting the polynucleotides into the cells or glioma (see, e.g., Acsadi G et al, Nature 332:815-18 (1991); Wolff J A et al., Science 247:1465-68 (1990)).
  • a delivery apparatus e.g., a "gene gun”
  • Such an apparatus is commercially available (e.g., from Bio-Rad Laboratories, Hercules, Calif.).
  • gliomas are contacted with NFlA inhibitors via direct injection of the NFIA inhibitor to the glioma.
  • Cationic Lipids Naked polynucleotides can be introduced into cells in vivo by complexing the polynucleotides with cationic lipids or encapsulating the polynucleotides in cationic liposomes.
  • Suitable cationic lipid formulations include N-[-l-(2,3-dioleoyloxy)propyl]N,N,N-triethylammonmm chloride (DOTMA) and a 1 :1 molar ratio of l,2-dimyristyloxy-propyl-3-dimethylhydroxyethyIammonium bromide (DMRIE) and dioleoyl phosphatidylethanol amine (DOPE) (see e.g., Logan J J et al., Gene Ther. 2:38-49 (1995); San H et al., Hum. Gene Ther. 4:781-88 (1993)).
  • DOTMA N-[-l-(2,3-dioleoyloxy)propyl]N,N,N-triethylammonmm chloride
  • DMRIE dioleoyl phosphatidylethanol amine
  • DOPE dioleoyl phosphatidyl
  • Naked polynucleotides can also be introduced into cells in vivo by complexing the polynucleotides to a cation, such as polylysine, which is coupled to a ligand for a cell-surface receptor (see, e.g., Wu G Y and Wu C H, J. Biol. Chem. 263:14621-24 (1988); Wilson J M et al., J. Biol. Chem. 267:963-67 (1992); and U.S. Pat. No. 5,166,320). Binding of the polymicleotide-ligand complex to the receptor facilitates uptake of the polynucleotide by receptor-mediated endocytosis.
  • a cation such as polylysine
  • a polynucleotide-ligand complex linked to adenovirus capsids which naturally disrupt endosomes, thereby releasing material into the cytoplasm can be used to avoid degradation of the complex by intracellular Iysosomes (see, e.g., Curiel D T et al., Proc. Natl. Acad. Sci, USA 88:8850-54 (1991); Cristiano R J et al., Proc. Natl. Acad. Sci. USA 90:2122-26 (1993)).
  • gliomas are contacted with NFIA inhibitors via receptor mediated uptake of the NFIA inhibitor.
  • Retroviruses Modified retroviruses are well characterized for use in gene transfer for gene therapy purposes (for a review, see Miller A D, Blood 76:271-78 (1990)).
  • a recombinant retrovirus can be constructed having a nucleotide sequence of interest incorporated into the retroviral genome. Additionally, portions of the retroviral genome can be removed to render the retrovirus replication defective. The replication defective retrovirus is then packaged into virions which can be used to infect a target cell through the use of a helper virus by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausube] F M et al.
  • retroviruses examples include pL. ⁇ , pZIP, pWE, and pEM which are well known to those skilled in the art.
  • suitable packaging virus lines include ⁇ pCrip, ⁇ pCre, ⁇ p2 and ⁇ pAm.
  • Retroviruses have been used to introduce a variety of genes into many different cell types, including epithelial cells, endothelial cells, lymphocytes, myoblasts, hepatocytes, bone marrow cells, in vitro and/or in vivo (see, e.g., Eglitis M A et al., Science 230: 1395-98 (1985); Danos O and Mulligan R C, Proc. Natl. Acad. Sci. USA 85:6460-64 (1988); Wilson J M et al., Proc. Natl. Acad. Sci. USA 85:3014-18 (1988); Armentano D et al., Proc. Natl. Acad. Sci.
  • Retroviral vectors require target cell division in order for the retroviral genome (and foreign nucleic acid inserted into it) to be integrated into the host genome to stably introduce nucleic acid into the cell. Thus, it may be necessary to stimulate replication of the target cell, hi some embodiments, retroviruses are used to deliver NFIA inhibitors to gliomas.
  • Adenoviruses The genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle (see, e.g., Berkner K L, Biotechniques 6:616-29 (1988); Rosenfeld M A et al., Science 252:431-34 (1991); and Rosenfeld M A et al., Cell 68:143-55 (1992)).
  • Suitable adenoviral vectors derived from the adenovirus strain Ad type 5 dl324 or other strains of adenovirus are well known to those skilled in the art.
  • Recombinant adenoviruses are advantageous in that they do not require dividing cells to be effective gene delivery vehicles and can be used to infect a wide variety of cell types, including airway epithelium (Rosenfeld M A et al., Cell 68:143-55 (1992)), endothelial cells (Lemarchand P et al., Proc. Natl. Acad. Sci. USA 89:6482-86 (1992)), hepatocytes (Herz J and Gerard R D, Proc. Natl. Acad. Sci. USA 90:2812-16 (1993)), and muscle cells (Quantin B et al., Proc. Natl. Acad. Sci. USA 89:2581-84 (1992)).
  • introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA).
  • the carrying capacity of the adenoviral genome for foreign DNA is large ⁇ up to 8 kilobases) relative to other gene delivery vectors (Berkner K L et al., supra; Haj-Ahmad Y and Graham F L, J. Virol. 57:267- 74 (1986)).
  • adenoviruses are used to deliver NFIA inhibitors to a glioma.
  • Adeno-associated virus is a naturally occurring defective virus that requires another virus, such as an adenovirus or a herpes virus, as a helper virus for efficient replication and a productive life cycle (for a review, see Muzyczka N, Curr. Top. Microbiol. Immunol. 158:97-129 (1992)). It is also one of the few viruses that may integrate its DNA into non-dividing cells, and exhibits a high frequency of stable integration (see, e.g., Flotte T R et al., Am. J. Respir. Cell. MoI. Biol. 7:349-56 (1992); Samulski R J et al., J. Virol.
  • AAV vector such as that described in Tratschin J D et al., MoI. Cell. Biol. 5:3251-60 (1985), can be used to introduce the polynucleotides into cells.
  • a variety of nucleic acids have been introduced into different cell types using AAV vectors (see, e.g., Hermonat P L and Muzyczka N, Proc. Natl. Acad. Sci.
  • adeno-associated viruses are used to deliver NFIA inhibitors to gliomas.
  • Lentivirus refers to a genus of retroviruses that are capable of infecting dividing and non-dividing cells.
  • lentiviruses include HIV (human immunodeficiency virus: including HIV type 1 , and HIV type 2), the etiologic agent of the human acquired immunodeficiency syndrome (AIDS); visna-maedi, which causes encephalitis (visna) or pneumonia (maedi) in sheep, the caprine arthritis-encephalitis virus, which causes immune deficiency, arthritis, and encephalopathy in goats; equine infectious anemia virus, which causes autoimmune hemolytic anemia, and encephalopathy in horses; feline immunodeficiency virus (FIV), which causes immune deficiency in cats; bovine immune deficiency virus (BIV), which causes lymphadenopathy, lymphocytosis, and possibly central nervous system infection in cattle; and simian immunodeficiency virus (SIV).
  • HIV human immunodeficiency virus: including HIV
  • an antisense NFIA polynucleotide e.g., an NFIA shRNA oligonucleotide or the like
  • NFIA antisense compounds are provided in a modified lentivirus expression vector, such as those described in U.S. Patent No. 7,195,916, herein incorporated by reference in its entirety.
  • Antisense polynucleotides may be produced from a heterologous expression cassette in a transfectant cell or transgenic cell.
  • the antisense polynucleotides may comprise soluble oligonucleotides that are administered to the external milieu, either in the culture medium in vitro or in the circulatory system or in interstitial fluid in vivo. Soluble antisense polynucleotides present in the external milieu have been shown to gain access to the cytoplasm and inhibit translation of specific mRNA species.
  • NFIA inhibitors can be administered to subjects in a biologically compatible form suitable for pharmaceutical administration in vivo to suppress NFIA activity.
  • biologically compatible form suitable for administration in vivo is meant a form of the NFIA inhibitor to be administered in which any toxic effects are outweighed by the therapeutic effects of the inhibitor.
  • subject is intended to include living organisms in which an immune response can be elicited, for example, mammals.
  • Administration of NFIA inhibitors as described herein can be in any pharmacological form including a therapeutically active amount of an agent alone or in combination with a pharmaceutically acceptable carrier.
  • a therapeutically active amount of the NFIA inhibitors disclosed herein is defined as an amount effective, at dosages and for periods of time necessary, to achieve the desired result.
  • a therapeutically active amount of a NFIA inhibitor may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of peptide to elicit a desired response in the individual. Dosage procedures may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily, or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
  • compositions of the present invention can be administered by any suitable route known in the art including, for example, intravenous, subcutaneous, intramuscular, transdermal, intrathecal, or intracerebral or administration to cells in ex vivo treatment protocols. Administration can be either rapid as by injection or over a period of time as by slow infusion or administration of slow release formulation.
  • NFIA inhibitors can be in the form of pharmaceutical preparations. Such preparations are made in a manner well known in the pharmaceutical art. One preferred preparation utilizes a vehicle of physiological saline solution, but it is contemplated that other pharmaceutically acceptable carriers such as physiological concentrations of other non-toxic salts, five percent aqueous glucose solution, sterile water or the like may also be used. As used herein "pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art.
  • any conventional media or agent is incompatible with the active compound, use thereof in the therapeutic compositions is contemplated.
  • Supplementary active compounds can also be incorporated into the compositions. It may also be desirable that a suitable buffer be present in the composition.
  • Such solutions can, if desired, be lyophilized and stored in a sterile ampoule ready for reconstitution by the addition of sterile water for ready injection.
  • the primary solvent can be aqueous or alternatively nonaqueous.
  • NFlA inhibitors can also be incorporated into a solid or semi-solid biologically compatible matrix which can be implanted into tissues requiring treatment.
  • the carrier can also contain other pharmaceutically-acceptable excipients for modifying or maintaining the pH, osmolality, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor of the formulation.
  • the carrier may contain still other pharmaceutically-acceptable excipients for modifying or maintaining release or absorption or penetration across the blood-brain barrier.
  • excipients are those substances usually and customarily employed to formulate dosages for parenteral administration in either unit dosage or multi-dose form or for direct infusion by continuous or periodic infusion.
  • Dose administration can be repeated depending upon the pharmacokinetic parameters of the dosage formulation and the route of administration used.
  • the NFIA inhibitors are formulated for oral administration. Such formulations are preferably encapsulated and formulated with suitable carriers in solid dosage forms.
  • suitable carriers, excipients, and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, gelatin, syrup, methyl cellulose, methyl- and propylhydroxybenzoates, talc, magnesium, stearate, water, mineral oil, and the like.
  • the formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents, or flavoring agents.
  • the compositions may be formulated so as to provide rapid, sustained, or delayed release of the active ingredients after administration to the patient by employing procedures well known in the art.
  • the formulations can also contain substances that diminish proteolytic degradation and/or substances which promote absorption such as, for example, surface active agents.
  • NFIA inhibitors can be formulated for parenteral delivery, in some embodiments, parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the NFIA inhibitor and the particular therapeutic effect to be achieved and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
  • the specific dose can be readily calculated by one of ordinary skill in the art, e.g., according to the approximate body weight or body surface area of the patient or the volume of body space to be occupied. The dose will also be calculated dependent upon the particular route of administration selected. Further refinement of the calculations necessary to determine the appropriate dosage for treatment is routinely made by those of ordinary skill in the art. Such calculations can be made without undue experimentation by one skilled in the art in light of the activity disclosed herein in assay preparations of target cells. Exact dosages are determined in conjunction with standard dose- response studies.
  • the amount of the composition actually administered will be determined by a practitioner, in the light of the relevant circumstances including the condition or conditions to be treated, the choice of composition to be administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the chosen route of administration.
  • Toxicity and therapeutic efficacy of NFlA inhibitors can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, for determining the LD 50 ⁇ the dose lethal to 50% of the population) and the ED 5O ⁇ the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 /ED 50 .
  • NFIA inhibitors which exhibit large therapeutic indices are preferred. While NFIA inhibitors that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such inhibitors to the gliomas in order to minimize potential damage to non- gliomal cells and, thereby, reduce side effects.
  • NFIA inhibitors lie preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any NFIA inhibitor used in the methods described herein, the therapeutically effective dose can be estimated initially from cell culture assays. [00851 The effectiveness of a NFIA inhibitor can be determined by a screening assay as described herein, such as determining the level of NIFA protein in gliomas, determining the tumor size of gliomas, or the like.
  • some embodiments provide for the use of a NFIA inhibitor disclosed herein in the manufacture of a medicament for the reduction of glioma size, or for the treatment of glioma.
  • a subject that has, or is at risk of developing a glioma is identified by routine diagnostic methods, e.g., with neurological exams to check vision, hearing balance, coordination, reflexes, and the like, imaging techniques such as MRI (with or without contrast enhancements), CT scans, and the like.
  • subjects identified as having, or at risk of developing, a glioma is administered a therapeutically effective amount of an NFIA inhibitor, as described herein.
  • the subject's glioma is contacted with an NFIA inhibitor.
  • the size and progression of the subject's glioma is monitored before and after treatment with the NFIA inhibitor (e.g., with MRI, CT scans, or the like).
  • oligo design is as follows: [0092] Forward 5' ACC G(N20) TTCAAGAGA (20N)C
  • Oligonucleotides were synthesized using routine solid phase synthesis methods. The oligos were synthesized, annealed, kinase treated and cloned into a Bluescript- hU6 plasmid (a gift from D. Baltimore) (Qin et al. (2003) Proc. Nat. Acad. Sci. USA 100(1): 183- 188)) digested with Bbsl and HindIIL Clones were sequence verified, and the hU6-shRNAi cassette was shuttled into a lentiviral vector (FG-12/GFP (Qin et al., 2003).
  • Bluescript- hU6 plasmid a gift from D. Baltimore
  • Bbsl and HindIIL Clones were sequence verified, and the hU6-shRNAi cassette was shuttled into a lentiviral vector (FG-12/GFP (Qin et al., 2003).
  • NFIA antisense oligonucleotide sequence cNFIA resulted in complete knockdown of endogenous NFIA protein:
  • mutant NFIA antisense polynucleotide As a control, a mutant NFIA antisense polynucleotide was generated, which has four base changes (underlined) compared to SEQ ID NO: 1, such that the antisense oligonucleotide would not anneal to and inhibit the endogenous NFIA transcript.
  • the lentiviral sh-RNAi DNA is co-transfected using routine methods into 293T cells along with DNA constructs that provide the necessary genetic components for efficient viral generation in trans. After transfection the supernatant is collected and concentrated. The resultant supernatant served as the viral substrate utilized in the experiments discussed below.
  • NFIA shRNA constructs can be used to reduce NFIA transcription in glioma cells.
  • theU87 cell line was transduced with the lentiviral NFIA-shRNAi viral supernatant, (described in Example 1) lentiviral NFIA-shRNAi-mutant viral supernatant (described in Example 1), or was left untreated.
  • NFIA antisense inhibitors reduce growth of NFIA in gliomal cells, and demonstrate that NFlA antisense inhibitors are useful for the treatment of glioma.
  • NFlA antisense inhibitors reduce the growth of gliomal tumors
  • U87 gliomal cells were transduced with the lentiviral NFIA-shRNAi viral supernatant, lentiviral NFIA-shRNAi-mutant viral supernatant, or a lentilviral NFIA cDNA viral supernatant, as described in Example 1.
  • Approximately 2.5x10 5 cells were injected intracranially into mice, and tumor formation and growth was monitored using MRI.
  • transgenic NFIA mice (described in Neves et al Proc. Nat. Acad. Sci. USA (1999) 96(21): 11946-11951) was used to generate NFIA +/- and NFIA -/- neurospheres from E 11.5 telencephelon. Neurospheres were transduced with retroviruses expressing SV40 Large T antigen and constitutively activated EGFR-vIO. Expression of SV40 Large T antigen and EGFR-vIIl were validated via immunocytochemistry (not shown).
  • NFIA +/-; SV40:EGFR-vIII and NFIA -;SV40;EGFR- vlll neurospheres were injected subcutaneously into the necks of SCID mice. Six weeks post-injection, tumors were harvested and analyzed. The results are presented in Table 1.
  • This example illustrates the reduction of the size of a gliomal tumor in a subject.
  • a subject is identified as having, or at risk of developing a glioma such as an astrocytoma using routine diagnostic methods.
  • the subject is administered a daily dose of an NFIA inhibitor, such that the NFIA inhibitor contacts the glioma (e.g., by injection of the NFlA inhibitor into the glioma).
  • the daily dose of the NFIA inhibitor can, in some cases, range from about 0.01 ⁇ g/kg to about 1 mg/kg of subject body weight or more per day, depending on the factors mentioned above.
  • the dose of the NFIA inhibitor ranges from about 10 ⁇ g/kg/day to about 100 ⁇ g/kg/day.
  • the appropriate dosage and treatment regimen can be readily determined by the skilled artisan based on a number of factors, including but not limited to the nature of the NFIA inhibitor, and the subject's disease state (e.g., the size and/or stage of the glioma), or condition.
  • Levels of NFIA within the glioma can be determined before and after administration of the NFIA inhibitor.
  • the glioma tumor size can be determined before and after administration of the NFIA inhibitor.
  • This example illustrates the treatment of an astrocytoma in a subject.
  • a subject is identified as having, or at risk of developing an astrocytoma using routine diagnostic methods.
  • the subject is administered a daily dose of an NFIA inhibitor, such that the NFlA inhibitor contacts the astrocytoma (e.g., by injection of the NFIA inhibitor into the astrocytoma).
  • the daily dose of the NFIA inhibitor can, in some cases, range from about 0.01 ⁇ g/kg to about 1 mg/kg of subject body weight or more per day, depending on the factors mentioned above.
  • the dose of the NFlA inhibitor ranges from about 10 ⁇ g/kg/day to about 100 ⁇ g/kg/day.
  • the appropriate dosage and treatment regimen can be readily determined by the skilled artisan based on a number of factors, including but not limited to the nature of the NFIA inhibitor, and the subject's disease state (e.g., the size and/or stage of the tumor), or condition.
  • Levels of NFIA within the astrocytoma can be determined before and after administration of the NFIA inhibitor.
  • the astrocytoma tumor size can be determined before and after administration of the NFIA inhibitor.

Abstract

Disclosed herein are compositions comprising NFIA inhibitors, as well as methods of using the same to treat glioma and astrocytomas.

Description

NFIA IN GLIAL FATE DETERMINATION, GLIOMA THERAPY AND ASTROCYTOMA TREATMENT
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims priority to U.S. Provisional Application Serial No. 60/994,753, filed on September 21, 2007, by Anderson et al. and entitled "NFIA IN GLIAL FATE DETERMINATION, GLIOMA THERAPY AND ASTROCYTOMA TREATMENT WITH RNAi," which is hereby expressly incorporated by reference in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED R&D
[0002] This invention was made with Government support under Grant No. ROl - NS23476, awarded by the National Institutes of Health. The Government has certain rights in this invention.
PARTIES OF JOINT RESEARCH AGREEMENT
[0003] This invention was made as a result of activities undertaken within the scope of a joint research agreement between the California Institute of Technology and Children's Hospital, Los Angeles.
BACKGROUND OF THE INVENTION Field of the Invention
[0004] Embodiments disclosed herein relate to molecular medicine, and in particular to compositions and methods that can be used for glioma and astrocytoma therapy and treatment. Description of the Related Art
[0005] Gliomas, tumors derived from glia cells of the central nervous system (CNS) account for approximately 30% of all primary brain tumors in adults. The median survival rate is about 12 months. Gliomas are usually treated by surgery, chemotherapy, radiotherapy and/or fractionated stereotype radio surgery.
[0006] Gliomas are heterogeneous in their cellular content and can be divided into groups of astrocytomas, anablastic astrocytomas and glioblastoma multiformes. Traditional approaches to therapeutic intervention have relied upon either surgery, chemotherapy, or radiotherapy. Recent advances in molecular genetics have revealed many genetic mutations and associated signaling pathways that may play a causative role in the generation of gliomas. While these advances have provided numerous candidate pathways that can be utilized in the development of rational therapy rooted in the biology of the disease, it remains to be seen whether such approaches will come to fruition. As such, there is a continued need for gliomal therapies.
[0007] The Nuclear Factor I (NFI) family of genes are glial fate determinants in the developing central nervous system. Interestingly, NFIA is also expressed in all grades of human astroctyoma, the most deadly form of glioma. Described herein is Applicants' discovery that NFIA plays a causative role in glioma foπnation and Applicants' discovery of therapeutics based on the elucidation of NFIA' s role in glioma formation.
SUMMARY OF THE INVENTION
[0008] Provided herein are methods and compositions for glioma therapy and treatment. Some embodiments provided herein relate to methods for reducing the size of a glioma. Some embodiments relate to methods of arresting or slowing the development or progression of gliomal tumors. In some embodiments, the gliomas can be astrocytomas.
[0009] In some embodiments, the methods described herein include the steps of contacting a glioma with a therapeutically effective amount of an NFlA inhibitor. In some embodiments, the NFIA inhibitor can include an NFIA antisense polynucleotide, such as an NFIA short hairpin RNA (shRNA). In some embodiments, the shRNA NFlA inhibitor can target human NFIA. For example, in some embodiments, the NFIA inhibitor can be an NFIA shRNA that comprises the sequence of SEQ ID NO: 1. In some embodiments, the contacting step comprises contacting the glioma with a viral expression vector that encodes an NFIA antisense polynucleotide, such as a lentiviral expression vector.
[0010] In some embodiments, the NFIA inhibitor can be an antibody that specifically binds to NFIA.
[0011] Some embodiments provide an NFIA antisense polynucleotide. In some embodiments, the NFIA antisense polynucleotide comprises an isolated polynucleotide comprising the sequence of SEQ ID NO: 1. BRIEF DESCRIPTION OF THE DRAWINGS
[0012] Figure IA is a bar graph showing the relative expression levels of NFIA in U87 glioma cells that were either untreated, treated with a mutant NFIA shRNA, or treated with an NFIA shRNA that targets human NFIA.
[0013] Figure 2 is a graph showing the growth (as expressed in total cell number) of U87 glioma cells infected with either a viral construct encoding a human NFIA shRNA or a mutant version of the human NFIA shRNA, or that were not infected.
[0014] Figure 3 is a graph showing the size and frequency of tumors in mice receiving transplants of: U87 glioma cells transduced with a lentiviral expression vector encoding NFIA cDNA; untreated U87 cells, U87 cells transduced with either a lentiviral construct encoding a NFIA shRNA; or U87 cells transduced with a lentiviral construct encoding a mutant version of the NFIA shRNA.
[0015] Figures 4A-4D are images of MRIs of genetically modified U87 cells implanted into a mouse brain, 28 days post implantation. The arrows denote tumor growth. DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
[0016] The embodiments disclosed herein relate to the treatment of gliomas and are based, in part, on Applicants' discovery that Nuclear Factor IA (NFIA) plays a role in glioma formation.
[0017] Embodiments herein relate to methods for treating gliomas, for reducing the tumor size of gliomas, and for reducing the functional NFIA levels in gliomas with NFlA inhibitors. The term "NFIA inhibitor" refers to a compound or composition that reduces the functional NFlA levels in a target cell (e.g., a glioma cell). NFΪA inhibitors can function by inhibiting the expression of NFIA, for example by inhibiting transcription, translation, or processing of NFIA, or by inhibiting a functional activity of NFIA, such as DNA binding. NFIA inhibitors include, but are not limited to, antisense polynucleotides, antibodies, and small molecule inhibitors of NFIA. NFIA Antisense Polynucleotides
[0018] In some embodiments, an NFIA inhibitor can be an NFIA antisense polynucleotide. Examples of antisense polynucleotides useful in the embodiments disclosed herein include single-stranded DNAs and RNAs that bind to complementary target mRNA and inhibit translation and/or induce RNaseH-mediated degradation of the target transcript; siRNA oligonucleotides, which are short, double-stranded RNAs that activate the RNA interference (RNAi) pathway leading to target mRNA degradation; ribozymes, which are oligonucleotide-based endonucleases that are designed to cleave specific mRNA transcripts; and nucleic acid aptainers and decoys, which are non-naturally occurring oligonucleotides that bind to and block protein targets in a manner analogous to small molecule drugs.
[0019] As used herein, the teπn "oligonucleotide" refers to an oligomer or polymer of nucleic acids such as ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or mimetic s thereof. This term includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent intemucleoside (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions which function similarly. Modified or substituted oligonucleotides provide desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target and increased stability in the presence of nucleases.
[0020] In addition to antisense oligonucleotides that comprise only naturally occurring nucleobases, in some embodiments, the antisense oligonucleotides provided herein can be oligomeric oligonucleotide mimetics. Accordingly, in some embodiments, antisense compounds provided herein include oligonucleotides containing modified backbones or non- natural internucleoside linkages. Oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone.
[0021] In some embodiments, the antisense polynucleotides comprise oligomeric oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotri esters, aminoalkylphosphotri-esters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates, 5'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriest- ers, selenophosphates and borano- phosphates having normal 3'-5' linkages, T-S" linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3' to 3', 5' to 5' or 21 to 2' linkage. Oligonucleotides having inverted polarity can comprise a single 3' to 3' linkage at the 3'-most internucleotide linkage i,e. a single inverted nucleoside residue which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof). Various salts, mixed salts and free acid forms are also included.
[0022] Representative United States patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos.: 3,687,808; 4,469,863; 4,476,301 ; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321 ,131 ; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,823 ; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,194,599; 5,565,555; 5,527,899; 5,721,218; 5,672,697 and 5,625,050, each of which is herein incorporated by reference in its entirety.
[0023] In some embodiments, NFIA antisense polynucleotides can comprise modified oligonucleotide backbones that do not include a phosphorus atom. NFIA antisense polynucleotides can have backbones formed by short chain alkyl or cycloalkyl intemucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; riboacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts.
[0024] Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos.: 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; 5,792,608; 5,646,269 and 5,677,439, certain of which are commonly owned with this application, and each of which is herein incorporated by reference. [0025] In some embodiments, NFIA antisense polynucleotides can comprise mimetics wherein both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with non-naturally occurring groups. In some embodiments, the base units are maintained for hybridization with an appropriate target polynucleotides. For example, some embodiments provide NFlA antisense polynucleotides that comprise a peptide nucleic acid (PNA). In PNA compounds, the sugar-backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative United States patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos.: 5,539,082; 5,714,331 ; and 5,719,262, each of which is herein incorporated by reference in its entirety. Further teaching of PNA compounds can be found in Nielsen et al., Science, 1991, 254, 1497-1500.
[0026] In some embodiments, NFIA antisense oligonucleotides can have phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular -CH2-NH-O-CH2-, -CH2-N(CHs)-O-CH2- [known as a methylene (methylimino) or MMI backbone], -CH2-O-N(CH3)-CH2-, -CH2-N(CH3J-N(CH3)- CH2-- and — O— N(CH3)- CH2- CH2- [wherein the native phosphodi ester backbone is represented as -0-P-O-CH2-] of the above referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above referenced U.S. Pat. No. 5,602,240. Other mimetics that are present in some embodiments of the antisense polynucleotides disclosed herein include morpholino backbone structures described in U.S. Pat. No. 5,034,506, herein incorporated by reference in its entirety.
[0027] In some embodiments, the NFIA antisense polynucleotides can contain one or more substituted sugar moieties. In some embodiments, the antisense oligonucleotides disclosed herein comprise one of the following at the T position: OH; F; O— , S-, or N-alkyl; 0—, S-, or N-alkenyl; O— , S— or N-alkynyl; or O-alkyl-0-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C] to C|0 alkyl or C2 to Ci0 alkenyl and alkynyl. In some embodiments, the antisense oligonucleotides can include one of the following groups at the T position: O[(CH2)nO]mCH3, O(CH2)nOCH3, O(CH2)nNH2, O(CH2)nCH3, O(CH2)nONH2, and 0(CH2)nON[(CH2)nCH3)]2, where n and m are from 1 to about 10. In some embodiments, the antisense oligonucleotides comprise one of the following at the 2' position: Ci to C1O lower alkyl, substituted lower alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH5 SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties. In some embodiments, the antisense oligonucleotides include T- methoxyethoxy (2'-0-CH2CH2OCH3, also known as 2'-O-(2-methoxyethyl) or 2'-MOE) (Martin et al., HeIv. Chim. Acta, 1995, 78, 486-504) i.e., an alkoxyalkoxy group. In some embodiments, the antisense polynucleotides or oligonucleotides disclosed herein include T- dim ethyl aminooxyethoxy, i.e., a O(CH2)2ON(CH3)2 group, also known as 2'-DMAOE, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-O-dimethylaminoethoxyethyl or 2'-DMAEOE), i.e., 2'-O~CH2-O-CH2-N(CH2)2, also described in examples herein below.
[0028] In some embodiments, the NFIA antisense polynucleotides and oligonucleotides can include Locked Nucleic Acids (LNAs) in which the 2'-hydroxyl group is linked to the 3' or 4' carbon atom of the sugar ring thereby forming a bicyclic sugar moiety. The linkage can be a methelyne (-CH2- )n group bridging the T oxygen atom and the 4' carbon atom wherein n is 1 or 2. LNAs and preparation thereof are described in WO 98/39352 and WO 99/14226, each of which is herein incorporated by reference in its entirety.
[0029] In some embodiments, the NFIA antisense polynucleotides and oligonucleotides can include 2'-methoxy (2'-0-CH3), 2'-aminopropoxy (T- OCH2CH2CH2NH2), 2'-a!lyl (2'-CH2-CH.dbd.CH2), 2'-O-aHyl (2'-O-CH2-CH.dbd.CH2- ) and 2'-fluoro (2'-F). The 2 '-modification may be in the arabino (up) position or ribo (down) position. In some embodiments, the 2'-arabino modification is 2'-F. Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'- 5 linked oligonucleotides and the 5' position of 5' terminal nucleotide. Oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative United States patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos.: 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811 ; 5,576,427; 5,591 ,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; 5,792,747; and 5,700,920, certain of which are commonly owned with the instant application, and each of which is herein incorporated by reference in its entirety.
|0030] Oligonucleotides may also include nucleobase (often referred to in the art simply as "base") modifications or substitutions. As used herein, "unmodified" or "natural" nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-amino adenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouraciI, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyI and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7 -methyl guanine and 7-methyladenine, 2-F-adenine, 2-amino- adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine. Further modified nucleobases include tricyclic pyrimidines such as phenoxazine cytidine(lH-pyrimido[5,4-b][l,4]benzoxazi- n-2(3H)-one), phenothiazine cytidine (lH-pyrimido[5,4-b][l,4]benzothiazin- 2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g. 9-(2-ammoethoxy)-H-pyrimido[5,4-b][I,4]benzoxazin-2(3H)- one), carbazole cytidine (2H-pyrimido[4,5-b]indol-2-one), pyridoindole cytidine (H- pyrido[3',2':4,5]pyrrolo[2,3-d]pyrimidin-2-one). Modified nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. L, ed. John Wiley & Sons, 1990, those disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y. S., Chapter 15, Antisense Research and Applications, pages 289-302, Crooke, S. T. and Lebleu, B. ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2- aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.20C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., eds., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are presently preferred base substitutions, even more particularly when combined with 2'-O-methoxyethyl sugar modifications.
[0031] Representative United States patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Pat. No. 3,687,808, as well as U.S. Pat. Nos.: 4,845,205; 5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; 5,645,985; 5,830,653; 5,763,588; 6,005,096; and 5,681 ,941, each of which is herein incorporated by reference, and U.S. Pat. No. 5,750,692, which is commonly owned with the instant application and also herein incorporated by reference.
[0032] In some embodiments, the NFIA antisense oligonucleotides can be chemically linked to one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide. For example, in some embodiments, the NFlA antisense oligonucleotides can include conjugate groups covalently bound to functional groups such as primary or secondary hydroxyl groups. Exemplary conjugate groups include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers, and groups that reduce the anionic charge of the polynucleotides, thereby enhancing transport across cellular membranes. Conjugate groups include cholesterols, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes. Groups that enhance the pharmacodynamic properties, in the context of the NFlA antisense oligonucleotides disclosed herein, include groups that improve oligomer uptake, enhance oligomer resistance to degradation, and/or strengthen sequence-specific hybridization with RNA. Groups that enhance the pharmacokinetic properties, in the context of this invention, include groups that improve oligomer uptake, distribution, metabolism or excretion. Representative conjugate groups are disclosed for example in International Patent Application PCTYUS92/09196, filed Oct. 23, 1992, and PCT/US2007/003641, filed Feb. 9, 2007, the entire disclosures of which are incorporated herein by reference in their entireties.
[0033] Other exemplary conjugate moieties useful in the embodiments disclosed herein include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Let., 1994, 4, 1053-1060), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660, 306-309; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3, 2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20, 533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J., 1991, 10, 11 1 1-1118; Kabanov et al., FEBS Lett., 1990, 259, 327-330; Svinarchuk et al., Biochimie, 1993, 75, 49-54), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethyl- ammonium l,2-di-0-hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654; Shea et al., Nucl. Acids Res., 1990, 18, 3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969- 973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923-937. Oligonucleotides of the invention may also be conjugated to active drug substances, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fenbufen, ketoprofen, (S)-(+)-pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indomethicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic, for example. Oligonucleotide-drug conjugates and their preparation are described for example in U.S. patent application Ser. No. 09/334,130 (filed Jun. 15, 1999) which is incorporated herein by reference in its entirety. [0034] Representative United States patents that teach the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Pat. Nos.: 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,580,731 ; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,1 12,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941, certain of which are commonly owned with the instant application, and each of which is herein incorporated by reference.
[0035] In some embodiments, some or all positions in an NFIA antisense oligonucleotide or polynucleotide can be modified. In other embodiments, the NFlA antisense oligonucleotides do not contain any modified units. In still other embodiments, the NFIA antisense oligonucleotides disclosed herein are not uniformly modified. In some embodiments, more than one of the aforementioned modifications can be incorporated in a single NFIA antisense polynucleotide or oligonucleotide, or even at a single nucleoside within an oligonucleotide.
[0036] In some embodiments, the antisense compounds are chimeric. As used herein, the term "chimeric" antisense compounds or "chimeras," refer to antisense compounds, particularly oligonucleotides, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an oligonucleotide compound. These oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid. An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids. By way of example, RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene expression. Consequently, comparable results can often be obtained with shorter oligonucleotides when chimeric oligonucleotides are used, compared to phosphorothioate deoxyoligonucleotides hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
[0037] hi some embodiments, chimeric NFIA antisense compounds can be formed as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above. Such compounds have also been referred to in the art as hybrids or gapmers. Representative United States patents that teach the preparation of such hybrid structures include, but are not limited to, U.S. Pat. Nos.: 5,013,830; 5,149,797; 5,220,007; 5,256,775; 5,366,878; 5,403,711; 5,491,133; 5,565,350; 5,623,065; 5,652,355; 5,652,356; and 5,700,922, each of which is herein incorporated by reference in its entirety.
[0038] NFIA antisense compounds disclosed herein can be conveniently and routinely made through the well-known technique of solid phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, Calif.), BioAutomation (Irving, TX), or the like. Any other means for such synthesis known in the art may additionally or alternatively be employed. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and alkylated derivatives.
[0039] NFIA antisense polynucleotides and oligonucleotides provided herein include any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, NFlA antisense polynucleotides and oligonucleotides can include prodrugs and pharmaceutically acceptable salts of the antisense polynucleotides, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
|0040] As used herein, the term "prodrug" indicates a therapeutic agent that is prepared in an inactive form that is converted to an active form within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or conditions. By way of example only, some embodiments provide prodrug versions of the NFlA antisense polynucleotides and oligonucleotides prepared as SATE [(S-acetyl-2-thioethyl) phosphate] derivatives according to the methods disclosed in WO 93/24510 to Gosselin et al., published Dec. 9, 1993 or in WO 94/26764 and U.S. Pat. No. 5,770,713 to Imbach et al.
J0041] hi some embodiments, the NFIA antisense polynucleotides can be 5-10- mers, 10-15-mers, 15-20mers, 20-25 mers, 25-30-mers, 30-40-mers, 40-45-mers, 45-50-mers, 50-55-mers, 55-60-mers, 60-65-mers, 65-70-mers, 70-75-mers, 75-80-mers, 80-85-mers, 85- 90-mers, 90-95-mers, 95-100-mers, 100-120-mers, 120-140-mers, 140-160-mers, 160-180- mers, 180-200-mers, or greater, or any number in between, including full length genes or RNA transcripts thereof, hi some embodiments the NFIA antisense polynucleotide can be, for example, a 43-55-mer that forms a hairpin, wherein about 4-5 of the nucleotide/modified subunits form the turn.
[0042] In some embodiments, NFIA antisense polynucleotides target human NFlA. Several sequences of human NFlA are publicly available, and can be found, for example, under GENBANK™ accession numbers NM 005595 (SEQ ID NO: 3); NM 001 134673 (SEQ ID NO: 4). In some embodiments, NFIA antisense polynucleotides target NFIA from a different organism, such as a different mammal, such of mice, rats, rabbits, guinea pigs, dogs, cats, sheep, goats, cows, primates, such as monkeys, chimpanzees, and apes, and the like. The skilled artisan will readily appreciate, however, that antisense NFlA polynucleotides can be designed to target any NFIA sequence now known or discovered in the future
|0043] Accordingly, some embodiments provide NFIA antisense polynucleotides that comprise NFIA shRNA sequences. Exemplary NFIA shRNA antisense polynucleotides include, but are not limited to, polynucleotides that comprise, consist essentially of, or consist of at least about 20 consecutive nucleic acids of the nucleic acid sequence of SEQ ID NO: 1, or mimetics thereof. In some embodiments, the NFIA shRNA can have at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity to at least 20 nucleotides of the nucleic acid sequence of SEQ ID NO: 1. Sequence identity can be determined using mathematical algorithms known to those skilled in the art. A non-limiting example of a mathematical algorithm utilized for comparison of sequences is the algorithm of Karlin S and Altschul S F, Proc. Natl. Acad. Sci. USA 87:2264-68 (1990), modified as in Karlin S and Altschul S F, Proc. Natl. Acad. Sci. USA 90:5873-77 (1993). Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0) of Altschul S F et al., J. MoI. Biol. 215:403- 10 (1990). BLAST nucleotide searches can be performed with the NBLAST program score=100, wordlength=12 to obtain homologous nucleotide sequences. Another non- limiting algorithm useful for the comparison of sequences is the algorithm of Myers E W and Miller W, Comput. Appl. Biosci. 4:11-17 (1988). Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. Another example of a mathematical algorithm utilized for the alignment of nucleic acid sequences is the Wilbur-Lipman algorithm (Wilbur W J and Lipman D J3 Proc. Natl. Acad. Sci. USA 80:726-30 (1983)). When using the Wilbur-Lipman algorithm, a window of 20, gap penalty of 3, Ktuple of 3 can be used. Both The Wilbur-Lipman algorithm is incorporated, for example, into the MEGALIGN program (e.g., version 3.1.7) which is part of the DNASTAR sequence analysis software package.
[0044] Additional algorithms for sequence analysis are known in the art, and include ADVANCE and ADAM, described in Torelli A and Robotti C A, Comput. Appl. Biosci. 10:3-5 (1994); and FASTA, described in Pearson W R and Lipman D L Proc. Natl. Acad. Sci. USA 85:2444-48 (1988).
[0045] In addition to the sequences set forth above, the skilled artisan will appreciate that various other regions of the NFIA coding sequence can be used as siRNA antisense targets, and are useful in the methods described herein. Antisense targets are generally 15-25 bases long, and refer to the region of an mRNA to which an antisense compound hybridizes. Determination of regions of the NFIA coding sequence useful as NFIA inhibitors is NFIA siRNA antisense targets can be selected using routine methods known to those skilled in the art. For example, siRNA targets are routinely identified by scanning an mRNA sequence for AA dinucleotides and recording the 19 nucleotides immediately downstream of the AA. Other methods can also been used to select the siRNA targets. In one example, the selection of the siRNA target sequence is purely empirically determined (see, e.g., Sui G et al., Proc. Natl. Acad. Sci. USA 99:5515-20 (2002)), as long as the target sequence starts with GG and does not share significant sequence homology with other genes as analyzed by BLAST search. In another example, a more elaborate method is employed to select the siRNA target sequences. This procedure exploits an observation that any accessible site in endogenous mRNA can be targeted for degradation by synthetic oligodeoxyribonucleotide/RNase H method (see, e.g., Lee N S et al., Nature Biotechnol. 20:500-05 (2002)).
[0046] In another embodiment, an NFIA antisense hairpin siRNA expression cassette is constructed to contain the sequence corresponding to the target, followed by a short spacer, the antisense strand of the target, and 5-6 Ts as transcription terminator. The order of the sense and antisense strands within the siRNA expression constructs can be altered without affecting the gene silencing activities of the hairpin siRNA. Lm certain instances, the reversal of the order may cause partial reduction in gene silencing activities.
[0047] hi some embodiments, the length of nucleotide sequences that serve as each side of the stem of the hairpin siRNA expression cassette can range, for instance, from 19 to 29 or more. In some embodiments, the loop size can range from 3 to 23 nucleotides. The skilled artisan will appreciate that other lengths and/or loop sizes suitable for siRNAs can also be used. For example, in some embodiments, the stem of the hairpin can be greater than 29 bases long, e.g., 30-50, bases, or more. In some embodiments, the stem of the hairpin can be less than 29 bases, e.g. 5-28 bases. In some embodiments, the loop can be greater than 23 bases long, e.g., 24-50 bases.
{0048] In yet another embodiment, a 5' overhang in the hairpin siRNA construct can be used, provided that the hairpin siRNA provides the descired therapeutic benefit, or reduces the levels of functional NFIA as described herein. In one specific example, the 5' overhang includes about 6 nucleotide residues.
[0049] LQ still yet another embodiment, the target sequence for RNAi is a 21-mer sequence. The 5' end of the target sequence has the dinucleotide "NA", where "N" can be any base and "A" represents adenine. The remaining 19-mer sequence has a GC content of between 35% and 55%. Ln addition, the remaining 19-mer sequence does not include any four consecutive A or T (i.e., AAAA or TTTT), three consecutive G or C (i.e., GGG or CCC), or seven "GC" in a row. [0050] Additional criteria can also be used for selecting RNAi target sequences. For instance, the GC content of the remaining 19-mer sequence can be limited to between 45% and 55%. Moreover, any 19-mer sequence having three consecutive identical bases (i.e., GGG, CCC, TTT, or AAA) or a palindrome sequence with 5 or more bases may be excluded. Furthermore, the remaining 19-mer sequence can be selected to have low sequence homology to other genes. In one specific example, potential target sequences are searched by BLASTN against NCBFs human UniGene cluster sequence database. The human UniGene database contains non-redundant sets of gene-oriented clusters. Each UniGene cluster includes sequences that represent a unique gene. 19-mer sequences producing no hit to other human genes under the BLASTN search can be selected.
[0051] The effectiveness of the siRNA sequences, as well as any other RNAi sequence according to the embodiments disclosed herein, can be evaluated using various methods known in the art. For instance, an NFIA siRNA sequence of the present invention can be introduced into a cell that expresses the NF3A gene. The polypeptide or mRNA level of the NFIA gene in the cell can be detected. A substantial change in the expression level of the NFIA gene before and after the introduction of the siRNA sequence is indicative of the effectiveness of the siRNA sequence in suppressing the expression of the NFlA gene. In one specific example, the expression levels of other genes are also monitored before and after the introduction of the siRNA sequence. An siRNA sequence which has inhibitory effect on NFlA gene expression but does not significantly affect the expression of other genes can be selected. In another specific example, multiple siRNA or other RNAi sequences can be introduced into the same target cell. These siRNA or RNAi sequences specifically inhibit NFIA gene expression but not the expression of other genes. In yet another specific example, siRNA or other RNAi sequences that inhibit the expression of the NFIA gene and one or more additional genes can be used. Anti -NFIA Antibodies
[0052] In some embodiments, an NFIA inhibitor can be an anti-NFIA antibody. The term "antibody" is used in the broadest sense and specifically covers, for example, single anti-NFIA monoclonal antibodies (e.g. antagonist, and neutralizing antibodies), anti-NFIA antibody compositions with polyepitopic specificity, single chain anti-NFIA antibodies, and fragments of anti-NFIA antibodies (see below). The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. NFIA antibodies useful in the embodiments described herein are commercially available, or can be generated using routine methods known to those skilled in the art.
[0053] Routine screening can be used to identify anti-NFlA-antibodies that are useful in the methods described herein, e.g., to reduce the activity of NFIA, to reduce tumor size, to treat gliomas, etc. Such screening methods include those detailed in the examples herein.
[0054] Polyclonal anti-NFIA antibodies can be prepared as described above by immunizing a suitable subject with an NFIA immunogen. The anti-NFIA antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELlSA) using immobilized NFIA polypeptide. If desired, the antibody molecules directed against a NFIA polypeptide can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as protein A chromatography to obtain the IgG fraction. At an appropriate time after immunization, for example, when the anti-NFIA antibody titers are highest, antibody- producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler G and Milstein C, Nature 256:495-97 (1975) (see also, Brown J P et al., J. Immunol. 127:539- 46 (1981): Brown J P et al., J. Biol. Chem. 255:4980-83 (1980); Yeh M Y et al., Proc. Natl. Acad. Sci. USA 76:2927-31 (1979); Yeh M Y et al., Int. J. Cancer 29:269-75 (1982)), the more recent human B cell hybridoma technique (Kozbor D and Roder J C. Immunol. Today 4:72-79 (1983)), the EBV-hybridoma technique (Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96), or trioma techniques. The technology for producing monoclonal antibody hybridomas is well known (see generally R. H. Kenneth, in Monoclonal Antibodies: A New Dimension In Biological Analyses, Plenum Publishing Corp., New York, N.Y. (1980); Lerner E A, Yale J. Biol. Med., 54:387-402 (1981); Gefter M L et al., Somatic Cell Genet. 3:231-36 (1977)). Briefly, an immortal cell line (typically a myeloma) is fused to lymphocytes (typically splenocytes) from a mammal immunized with a Ccdc80 immunogen as described above, and the culture supernatants of the resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds specifically to a Ccdc80 polypeptide
[0055] Any of the many well known protocols used for fusing lymphocytes and immortalized cell lines can be applied for the purpose of generating an anti-NFIA monoclonal antibody (see, e.g., Galfre G et al., Nature 266:550-52 (1977); Geifer M L et ah, supra; Lerner E A, supra; Kenneth, Monoclonal Antibodies, supra). Moreover, the ordinary skilled worker will appreciate that there are many variations of such methods which also would be useful. Typically, the immortal cell line (e.g., a myeloma cell line) is derived from the same mammalian species as the lymphocytes. For example, murine hybridomas can be made by fusing lymphocytes from a mouse immunized with an immunogenic preparation of the present invention with an immortalized mouse cell line. Preferred immortal cell lines are mouse myeloma cell lines that are sensitive to culture medium containing hypoxanthine, aminopterin and thymidine ("HAT medium"). Any of a number of myeloma cell lines may be used as a fusion partner according to standard techniques, for example, the P3-NSl/l-Ag4-l , P3-x63-Ag8.653 or Sp2/0-Agl4 myeloma lines. These myeloma lines are available from the American Type Culture Collection (ATCC), Rockville, MD. Typically, HAT-sensitive mouse myeloma cells are fused to mouse splenocytes using polyethylene glycol ("PEG"). Hybridoma cells resulting from the fusion are then selected using HAT medium, which kills unfused and unproductively fused myeloma cells (unfused splenocytes die after several days because they are not transformed). Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind a NFLA molecule, for example, using a standard ELISA assay.
[0056] As an alternative to preparing monoclonal antibody-secreting hybridomas, a monoclonal anti-NFIA antibody can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with NFIA to thereby isolate immunoglobulin library members that bind a NFIA polypeptide. Kits for generating and screening phage display libraries are commercially available (e.g., the GE Healthcare Recombinant Phage Antibody System, Catalog No. 27-9400-01). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S. Pat. No. 5,223,409; WO 92/18619; WO 91/17271; WO 92/20791 ; WO 92/15679; WO 93/01288; WO 92/01047; WO 92/09690; WO 90/02809; Fuchs P et al., Biotechnology (N.Y.) 9:1370-72 (1991); Hay B N et al., Hum. Antibodies Hybridomas 3:81-85 (1992); Huse W D et al., Science 246:1275-81 (1989); Griffiths A D et al., EMBO J. 12:725-34 (1993); Hawkins R E et al., J. MoI. Biol. 226:889-96 (1992); Clarkson T et al., Nature 352:624-28 (1991); Gram H et al., Proc. Natl. Acad. Sci. USA 89:3576-80 (1992); Garrard L J et al., Biotechnology (N.Y.) 9: 1373-77 (1991); Hoogenboom H R et al, Nucleic Acids Res. 19:4133-37 (1991); Barbas C F et al., Proc. Natl. Acad. Sci. USA 88:7978-82 (1991); and McCafferty J et al., Nature 348:552-54 (1990).
|0057] Additionally, recombinant anti-NFIA antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, can be produced by recombinant DNA techniques known in the art, for example using methods described in WO 87/02671 ; EP 0 184 187; EP 0 171 496; EP 0 173 494; WO 86/01533; U.S. Pat. No. 4,816,567; EP 0 125 023; Better M et al., Science 240: 1041-43 (1988); Liu A Y et al., Proc. Natl. Acad. Sci. USA 84:3439-43 (1987); Liu A Y et al., J. Immunol. 139:3521-26 (1987); Sun L K et al., Proc. Natl. Acad. Sci. USA 84:214-18 (1987); Nishimura Y et al., Cancer Res. 47:999-1005 (1987); Wood C R et al., Nature 314:446-49 (1985); Shaw D R et al., J. Natl. Cancer Inst. 80:1553-59 (1988); Morrison S L, Science 229: 1202-07 (1985); U.S. Pat. No. 5,225,539; Verhocyan M et al., Science 239:1534-36 (1988); and Beidler C B et al., J. Immunol. 141 :4053-60 (1988).
[0058] In addition, humanized antibodies can be made according to standard protocols such as those disclosed in U.S. Pat. No, 5,565,332. In another embodiment, antibody chains or specific binding pair members can be produced by recombination between vectors comprising nucleic acid molecules encoding a fusion of a polypeptide chain of a specific binding pair member and a component of a replicable genetic display package and vectors containing nucleic acid molecules encoding a second polypeptide chain of a single binding pair member using techniques known in the art, for example, as described in U.S. Pat. No. 5,565,332; 5,871,907; or 5,733,743. [0059] In some embodiments, anti-NFIA antibodies that recognize intracellular NFIA can be used, e.g., intracellular^ to inhibit NFIA protein activity. The use of intracellular antibodies to inhibit protein function in a cell is known in the art (see e.g., Carlson J R3 MoI. Cell. Biol. 8:2638-46 (1988); Biocca S et al., EMBO J. 9:101-08 (1990); Werge T M et al., FEBS Lett. 274: 193-98 (1990); Carlson J R, Proc. Natl. Acad. Sci. USA 90:7427-28 (1993); Marasco W A et al., Proc. Natl. Acad. Sci. USA 90:7889-93 (1993); Biocca S et al., Biotechnology (N.Y.) 12:396-99 (1994); Chen S-Y et al., Hum. Gene Ther. 5:595-601 (1994); Duan L et al., Proc. Natl Acad. Sci. USA 91 :5075-79 (1994); Chen S-Y et al., Proc. Natl. Acad. Sci. USA 91 :5932-36 (1994); Beerli R R et al., J. Biol. Chem. 269:23931-36 (1994); Beerli R R et al., Biochem. Biophys. Res. Commun. 204:666-72 (1994); Mhashilkar A M et al., EMBO J. 14:1542-51 (1995); Richardson J H et al., Proc. Natl. Acad. Sci. USA 92:3137-41 (1995); WO 94/02610; and WO 95/03832).
[0060] In one embodiment, a recombinant expression vector is prepared which encodes the antibody chains in a form such that, upon introduction of the vector into a cell, the antibody chains are expressed by the cell as a functional antibody. For inhibition of NFlA activity according to the inhibitory methods of the invention, an intracellular antibody that specifically binds NFIA protein is preferably secreted from the cell.
[0061] To prepare an antibody expression vector, antibody light and heavy chain cDNAs encoding antibody chains specific for the target protein of interest, for example, Ccdc80, are isolated, typically from a hybridoma that secretes a monoclonal antibody specific for the NFIA protein. Hybridomas secreting anti-NFIA monoclonal antibodies, or recombinant anti-NFIA monoclonal antibodies, can be prepared as described above. Once a monoclonal antibody specific for NFlA protein has been identified (e.g., either a hybridoma- derived monoclonal antibody or a recombinant antibody from a combinatorial library), DNAs encoding the light and heavy chains of the monoclonal antibody are isolated by standard molecular biology techniques. For hybridoma derived antibodies, light and heavy chain cDNAs can be obtained, for example, by PCR amplification or cDNA library screening. For recombinant antibodies, such as from a phage display library, cDNA encoding the light and heavy chains can be recovered from the display package (e.g., phage) isolated during the library screening process. Nucleotide sequences of antibody light and heavy chain genes from which PCR primers or cDNA library probes can be prepared are known in the art. For example, many such sequences are disclosed in Kabat E A et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242 and in the "Vbase" human germline sequence database.
[0062] Once obtained, the antibody light and heavy chain sequences are cloned into a recombinant expression vector using standard methods. An antibody expression vector can encode an antibody in one of several different forms. For example, in one embodiment, the vector encodes full-length antibody light and heavy chains such that a full-length antibody is expressed. To inhibit NFIA activity in a cell, the expression vector encoding the anti-NFIA intracellular or extracellular antibody is introduced into the cell by standard transfection methods, as discussed herein.
{0063] Anti-NFIA antibodies that function as NFIA inhibitors can be identified by screening for a desired inhibitory activity, for example, by using the methods described in the examples and elsewhere in the disclosure. For example, anti-NFIA antibodies can be screened in vitro for their ability to slow or arrest growth of gliomas, to decrease NFIA expression in gliomal cells, inhibiot NFIA activity (e.g., inhibit transcription of NFIA- induced genes, etc),or the like. Pharmaceutical Compositions and Methods
[0064] In some embodiments, the methods disclosed herein (e.g., reduction of NFIA activity, treatment of gliomas, treatment of astrocytomas, etc.) involve contacting a glioma with an effective amount of an NFlA inhibitor. The terms "effective amount", "therapeutically effective amount", and "effective dosage" as used herein refer to the amount of an NFIA inhibitor that is effective to at least slow the rate of glioma tumor growth, slow or arrest the progression of glioma, or decrease glioma tumor size. Tumor growth and tumor size can be measured using routine methods known to those skilled in the art, including, for example, magnetic resonance imagine and the like. Gliomas can be subdivided based on the WHO grading system into grade HV3 with grade I reflecting a benign, highly differentiated tumor and grades H-IV marking increasing states of malignancy, and decreasing degrees of differentiation. In some embodiments, an "effective amount" of an NFIA inhibitor is an amount effective to result in a downgrading of a tumor, or an amount effective to slow or prevent the progression of a glioma to a higher grade.
[0065J In embodiments wherein the NFIA inhibitor is a antisense oligonucleotide, for example, a an "effective amount" can refer to the amount of NFIA inhibitor necessary to cause "knock-down" of NFIA expression relative to levels present following administration or expression of a non-targeting control RNA (e.g., a non-targeting control siRNA). Knockdown of expression of an amount including and between 50% and 100%, e.g., 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or more, is contemplated by embodiments herein. Knock-down can be assessed by measuring the mRNA levels using quantitative polymerase chain reaction (qPCR) amplification or by measuring protein levels by western blot or enzyme-linked immunosorbent assay (ELISA). Analyzing the protein level provides an assessment of both mRNA cleavage as well as translation inhibition. Further techniques for measuring knock-down include RNA solution hybridization, nuclease protection, northern hybridization, gene expression monitoring with a microarray, antibody binding, radioimmunoassay, and fluorescence activated cell analysis.
[0066] hi some embodiments, the methods disclosed herein include contacting a glioma or glioma target cell with an effective amount of an NFIA inhibitor. In embodiments wherein the NFIA inhibitor comprises a polynucleotide, (including recombinant expression vectors encoding NFIA antisense RNA, intracellular NFIA antibodies, or dominant negative NFIA inhibitors), the agents can be introduced into cells of the glioma using methods known in the art for introducing polynucleotides (e.g., DNA, RNA or the like) into cells in vivo. Examples of such methods encompass both non-viral and viral methods, including but not limited to the methods described below.
[00671 Direct Injection: Naked polynucleotides can be introduced into cells or glioma in vivo by directly injecting the polynucleotides into the cells or glioma (see, e.g., Acsadi G et al, Nature 332:815-18 (1991); Wolff J A et al., Science 247:1465-68 (1990)). For example, a delivery apparatus (e.g., a "gene gun") for injecting polynucleotides into cells in vivo can be used. Such an apparatus is commercially available (e.g., from Bio-Rad Laboratories, Hercules, Calif.). In some embodiments, gliomas are contacted with NFlA inhibitors via direct injection of the NFIA inhibitor to the glioma. [0068] Cationic Lipids: Naked polynucleotides can be introduced into cells in vivo by complexing the polynucleotides with cationic lipids or encapsulating the polynucleotides in cationic liposomes. Examples of suitable cationic lipid formulations include N-[-l-(2,3-dioleoyloxy)propyl]N,N,N-triethylammonmm chloride (DOTMA) and a 1 :1 molar ratio of l,2-dimyristyloxy-propyl-3-dimethylhydroxyethyIammonium bromide (DMRIE) and dioleoyl phosphatidylethanol amine (DOPE) (see e.g., Logan J J et al., Gene Ther. 2:38-49 (1995); San H et al., Hum. Gene Ther. 4:781-88 (1993)).
[0069] Receptor-Mediated DNA Uptake: Naked polynucleotides can also be introduced into cells in vivo by complexing the polynucleotides to a cation, such as polylysine, which is coupled to a ligand for a cell-surface receptor (see, e.g., Wu G Y and Wu C H, J. Biol. Chem. 263:14621-24 (1988); Wilson J M et al., J. Biol. Chem. 267:963-67 (1992); and U.S. Pat. No. 5,166,320). Binding of the polymicleotide-ligand complex to the receptor facilitates uptake of the polynucleotide by receptor-mediated endocytosis. A polynucleotide-ligand complex linked to adenovirus capsids which naturally disrupt endosomes, thereby releasing material into the cytoplasm can be used to avoid degradation of the complex by intracellular Iysosomes (see, e.g., Curiel D T et al., Proc. Natl. Acad. Sci, USA 88:8850-54 (1991); Cristiano R J et al., Proc. Natl. Acad. Sci. USA 90:2122-26 (1993)). In some embodiments, gliomas are contacted with NFIA inhibitors via receptor mediated uptake of the NFIA inhibitor.
}0070| Retroviruses: Modified retroviruses are well characterized for use in gene transfer for gene therapy purposes (for a review, see Miller A D, Blood 76:271-78 (1990)). A recombinant retrovirus can be constructed having a nucleotide sequence of interest incorporated into the retroviral genome. Additionally, portions of the retroviral genome can be removed to render the retrovirus replication defective. The replication defective retrovirus is then packaged into virions which can be used to infect a target cell through the use of a helper virus by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausube] F M et al. (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals. Examples of suitable retroviruses include pL.Ϊ, pZIP, pWE, and pEM which are well known to those skilled in the art. Examples of suitable packaging virus lines include ψpCrip, ψpCre, ψp2 and ψpAm. Retroviruses have been used to introduce a variety of genes into many different cell types, including epithelial cells, endothelial cells, lymphocytes, myoblasts, hepatocytes, bone marrow cells, in vitro and/or in vivo (see, e.g., Eglitis M A et al., Science 230: 1395-98 (1985); Danos O and Mulligan R C, Proc. Natl. Acad. Sci. USA 85:6460-64 (1988); Wilson J M et al., Proc. Natl. Acad. Sci. USA 85:3014-18 (1988); Armentano D et al., Proc. Natl. Acad. Sci. USA 87:6141-45 (1990); Huber B E et al., Proc. Natl. Acad. Sci. USA 88:8039-43 (1991); Ferry N et al., Proc. Natl. Acad. Sci. USA 88:8377-81 (1991); Chowdhury J R et al., Science 254:1802-05 (1991); van Beusechem V W et al., Proc. Natl. Acad. Sci. USA 89:7640-44 (1992); Kay M A et al., Hum. Gene Ther. 3:641-47 (1992); Dai Y et al., Proc. Natl. Acad. Sci. USA 89:10892-95 (1992); Hwu P et al., J. Immunol. 150:4104-15 (1993); U.S. Pat. No. 4,868,116; U.S. Pat. No. 4,980,286; WO 89/07136; WO 89/02468; WO 89/05345; and WO 92/07573). Retroviral vectors require target cell division in order for the retroviral genome (and foreign nucleic acid inserted into it) to be integrated into the host genome to stably introduce nucleic acid into the cell. Thus, it may be necessary to stimulate replication of the target cell, hi some embodiments, retroviruses are used to deliver NFIA inhibitors to gliomas.
[0071] Adenoviruses: The genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle (see, e.g., Berkner K L, Biotechniques 6:616-29 (1988); Rosenfeld M A et al., Science 252:431-34 (1991); and Rosenfeld M A et al., Cell 68:143-55 (1992)). Suitable adenoviral vectors derived from the adenovirus strain Ad type 5 dl324 or other strains of adenovirus (e.g., Ad2, Ad3, Ad7, etc.) are well known to those skilled in the art. Recombinant adenoviruses are advantageous in that they do not require dividing cells to be effective gene delivery vehicles and can be used to infect a wide variety of cell types, including airway epithelium (Rosenfeld M A et al., Cell 68:143-55 (1992)), endothelial cells (Lemarchand P et al., Proc. Natl. Acad. Sci. USA 89:6482-86 (1992)), hepatocytes (Herz J and Gerard R D, Proc. Natl. Acad. Sci. USA 90:2812-16 (1993)), and muscle cells (Quantin B et al., Proc. Natl. Acad. Sci. USA 89:2581-84 (1992)). Additionally, introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA). Moreover, the carrying capacity of the adenoviral genome for foreign DNA is large {up to 8 kilobases) relative to other gene delivery vectors (Berkner K L et al., supra; Haj-Ahmad Y and Graham F L, J. Virol. 57:267- 74 (1986)). Most replication-defective adenoviral vectors currently in use are deleted for all or parts of the viral El and E3 genes but retain as much as 80% of the adenoviral genetic material. In some embodiments, adenoviruses are used to deliver NFIA inhibitors to a glioma.
(0072) Adeno-Associated Viruses: Adeno- associated virus (AAV) is a naturally occurring defective virus that requires another virus, such as an adenovirus or a herpes virus, as a helper virus for efficient replication and a productive life cycle (for a review, see Muzyczka N, Curr. Top. Microbiol. Immunol. 158:97-129 (1992)). It is also one of the few viruses that may integrate its DNA into non-dividing cells, and exhibits a high frequency of stable integration (see, e.g., Flotte T R et al., Am. J. Respir. Cell. MoI. Biol. 7:349-56 (1992); Samulski R J et al., J. Virol. 63:3822-28 (1989); and McLaughlin S K et al., J. Virol. 62:1963-73 (1988)). Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate. Space for exogenous DNA is limited to about 4.5 kb. An AAV vector such as that described in Tratschin J D et al., MoI. Cell. Biol. 5:3251-60 (1985), can be used to introduce the polynucleotides into cells. A variety of nucleic acids have been introduced into different cell types using AAV vectors (see, e.g., Hermonat P L and Muzyczka N, Proc. Natl. Acad. Sci. USA 81 :6466-70 (1984); Tratschin J D et al., MoL Cell. Biol. 4:2072-81 (1985); Wondisford F E et al., MoI. Endocrinol. 2:32-39 (1988); Tratschin J D et al., J. Virol. 51:61 1- 19 (1984); and Flotte T R et al., J. Biol. Chem. 268:3781-90 (1993)). In some embodiments, adeno-associated viruses are used to deliver NFIA inhibitors to gliomas.
|0073| Lentiviruses: "Lentivirus" refers to a genus of retroviruses that are capable of infecting dividing and non-dividing cells. Several examples of lentiviruses include HIV (human immunodeficiency virus: including HIV type 1 , and HIV type 2), the etiologic agent of the human acquired immunodeficiency syndrome (AIDS); visna-maedi, which causes encephalitis (visna) or pneumonia (maedi) in sheep, the caprine arthritis-encephalitis virus, which causes immune deficiency, arthritis, and encephalopathy in goats; equine infectious anemia virus, which causes autoimmune hemolytic anemia, and encephalopathy in horses; feline immunodeficiency virus (FIV), which causes immune deficiency in cats; bovine immune deficiency virus (BIV), which causes lymphadenopathy, lymphocytosis, and possibly central nervous system infection in cattle; and simian immunodeficiency virus (SIV), which cause immune deficiency and encephalopathy in sub-human primates, hi some embodiments, the NFIA antisense oligonucleotides can be provided in an expression vector, such as a lentilviral vector. In some embodiments, an antisense NFIA polynucleotide (e.g., an NFIA shRNA oligonucleotide or the like), can be used to deliver the NFIA inhibitor to the glioma or target cell. In some embodiments, NFIA antisense compounds are provided in a modified lentivirus expression vector, such as those described in U.S. Patent No. 7,195,916, herein incorporated by reference in its entirety.
[0074] Antisense polynucleotides may be produced from a heterologous expression cassette in a transfectant cell or transgenic cell. Alternatively, the antisense polynucleotides may comprise soluble oligonucleotides that are administered to the external milieu, either in the culture medium in vitro or in the circulatory system or in interstitial fluid in vivo. Soluble antisense polynucleotides present in the external milieu have been shown to gain access to the cytoplasm and inhibit translation of specific mRNA species.
Administration of NFIA inhibitors
[0075] NFIA inhibitors can be administered to subjects in a biologically compatible form suitable for pharmaceutical administration in vivo to suppress NFIA activity. By "biologically compatible form suitable for administration in vivo" is meant a form of the NFIA inhibitor to be administered in which any toxic effects are outweighed by the therapeutic effects of the inhibitor. The term subject is intended to include living organisms in which an immune response can be elicited, for example, mammals. Administration of NFIA inhibitors as described herein can be in any pharmacological form including a therapeutically active amount of an agent alone or in combination with a pharmaceutically acceptable carrier. [0076] Administration of a therapeutically active amount of the NFIA inhibitors disclosed herein is defined as an amount effective, at dosages and for periods of time necessary, to achieve the desired result. For example, a therapeutically active amount of a NFIA inhibitor may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of peptide to elicit a desired response in the individual. Dosage regima may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily, or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
[0077] The therapeutic or pharmaceutical compositions of the present invention can be administered by any suitable route known in the art including, for example, intravenous, subcutaneous, intramuscular, transdermal, intrathecal, or intracerebral or administration to cells in ex vivo treatment protocols. Administration can be either rapid as by injection or over a period of time as by slow infusion or administration of slow release formulation.
[0078] NFIA inhibitors can be in the form of pharmaceutical preparations. Such preparations are made in a manner well known in the pharmaceutical art. One preferred preparation utilizes a vehicle of physiological saline solution, but it is contemplated that other pharmaceutically acceptable carriers such as physiological concentrations of other non-toxic salts, five percent aqueous glucose solution, sterile water or the like may also be used. As used herein "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the therapeutic compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions. It may also be desirable that a suitable buffer be present in the composition. Such solutions can, if desired, be lyophilized and stored in a sterile ampoule ready for reconstitution by the addition of sterile water for ready injection. The primary solvent can be aqueous or alternatively nonaqueous. NFlA inhibitors can also be incorporated into a solid or semi-solid biologically compatible matrix which can be implanted into tissues requiring treatment. [0079] The carrier can also contain other pharmaceutically-acceptable excipients for modifying or maintaining the pH, osmolality, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor of the formulation. Similarly, the carrier may contain still other pharmaceutically-acceptable excipients for modifying or maintaining release or absorption or penetration across the blood-brain barrier. Such excipients are those substances usually and customarily employed to formulate dosages for parenteral administration in either unit dosage or multi-dose form or for direct infusion by continuous or periodic infusion.
[0080] Dose administration can be repeated depending upon the pharmacokinetic parameters of the dosage formulation and the route of administration used.
[0081] In some embodiments, the NFIA inhibitors are formulated for oral administration. Such formulations are preferably encapsulated and formulated with suitable carriers in solid dosage forms. Some examples of suitable carriers, excipients, and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, gelatin, syrup, methyl cellulose, methyl- and propylhydroxybenzoates, talc, magnesium, stearate, water, mineral oil, and the like. The formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents, or flavoring agents. The compositions may be formulated so as to provide rapid, sustained, or delayed release of the active ingredients after administration to the patient by employing procedures well known in the art. The formulations can also contain substances that diminish proteolytic degradation and/or substances which promote absorption such as, for example, surface active agents.
[0082] In some embodiments, NFIA inhibitors can be formulated for parenteral delivery, in some embodiments, parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the NFIA inhibitor and the particular therapeutic effect to be achieved and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals. The specific dose can be readily calculated by one of ordinary skill in the art, e.g., according to the approximate body weight or body surface area of the patient or the volume of body space to be occupied. The dose will also be calculated dependent upon the particular route of administration selected. Further refinement of the calculations necessary to determine the appropriate dosage for treatment is routinely made by those of ordinary skill in the art. Such calculations can be made without undue experimentation by one skilled in the art in light of the activity disclosed herein in assay preparations of target cells. Exact dosages are determined in conjunction with standard dose- response studies. It will be understood that the amount of the composition actually administered will be determined by a practitioner, in the light of the relevant circumstances including the condition or conditions to be treated, the choice of composition to be administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the chosen route of administration.
[0083] Toxicity and therapeutic efficacy of NFlA inhibitors can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, for determining the LD50 {the dose lethal to 50% of the population) and the ED5O {the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. NFIA inhibitors which exhibit large therapeutic indices are preferred. While NFIA inhibitors that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such inhibitors to the gliomas in order to minimize potential damage to non- gliomal cells and, thereby, reduce side effects.
[0084] Data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of NFIA inhibitors lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any NFIA inhibitor used in the methods described herein, the therapeutically effective dose can be estimated initially from cell culture assays. [00851 The effectiveness of a NFIA inhibitor can be determined by a screening assay as described herein, such as determining the level of NIFA protein in gliomas, determining the tumor size of gliomas, or the like.
{0086] As such, some embodiments provide for the use of a NFIA inhibitor disclosed herein in the manufacture of a medicament for the reduction of glioma size, or for the treatment of glioma.
[0087] In some embodiments, a subject that has, or is at risk of developing a glioma is identified by routine diagnostic methods, e.g., with neurological exams to check vision, hearing balance, coordination, reflexes, and the like, imaging techniques such as MRI (with or without contrast enhancements), CT scans, and the like. In some embodiments, subjects identified as having, or at risk of developing, a glioma is administered a therapeutically effective amount of an NFIA inhibitor, as described herein. For example, in some embodiments, the subject's glioma is contacted with an NFIA inhibitor. In some embodiments, the size and progression of the subject's glioma is monitored before and after treatment with the NFIA inhibitor (e.g., with MRI, CT scans, or the like).
[0088] The following examples are presented in order to more fully illustrate some embodiments of the invention. They should, in no way be construed, however, as limiting the broad scope of the invention.
EXAMPLE 1 -NFIA SMALL HAIRPIN RNA REDUCES NFlA EXPRESSION IN
GLIOMA CELL LINES
[0089] This example describes the design of an exemplary NFIA shRNA.
[0090] To determine potential NFIA shRNA sequences, publicly available programs, including siRNA target designer, were used. The data generated by the designer programs was compared, and sequences that were identified by multiple programs were tested in vitro for silencing of cNFLA.
[0091] Briefly, the selected sequences were integrated into oligos containing a stem-loop sequence and engineered restriction enzyme sites to facilitate cloning. The format of the oligo design is as follows: [0092] Forward 5' ACC G(N20) TTCAAGAGA (20N)C
TTTTTA J
IΘ093] Reverse 3' - — C<N20) -AAGTTCTCT (20N)G
AAAAAAGCTT 5'
[0094] Oligonucleotides were synthesized using routine solid phase synthesis methods. The oligos were synthesized, annealed, kinase treated and cloned into a Bluescript- hU6 plasmid (a gift from D. Baltimore) (Qin et al. (2003) Proc. Nat. Acad. Sci. USA 100(1): 183- 188)) digested with Bbsl and HindIIL Clones were sequence verified, and the hU6-shRNAi cassette was shuttled into a lentiviral vector (FG-12/GFP (Qin et al., 2003).
[0095] The following NFIA antisense oligonucleotide sequence cNFIA resulted in complete knockdown of endogenous NFIA protein:
[0096] GAGGCACATGGAGAACTAAA (SEQ ID NO: 1).
[0097] As a control, a mutant NFIA antisense polynucleotide was generated, which has four base changes (underlined) compared to SEQ ID NO: 1, such that the antisense oligonucleotide would not anneal to and inhibit the endogenous NFIA transcript.
[0098] GA JGCACGTGAAGACGT AJ_A (SEQ ID NO : 2)
[0099] To generate the lentivirus, the lentiviral sh-RNAi DNA is co-transfected using routine methods into 293T cells along with DNA constructs that provide the necessary genetic components for efficient viral generation in trans. After transfection the supernatant is collected and concentrated. The resultant supernatant served as the viral substrate utilized in the experiments discussed below.
|0100] A second set of experiments, was performed to test whether the NFIA RNAi constructs described above efficiently reduced the level of NFIA expression in glioma cell lines. A small amount of the viral substrate described above was added to a plate of U87 human glioma cell lines. Efficient transduction of the lentivirus into the cells was measured by GFP expression. Forty-eight hours post infection, RNA was harvested from the cells and analyzed by quantitative PCR (qPCR).
[0101] U87 cells transfected with the NFlA-shRNAi described above exhibited an 80% reduction in the levels of NFIA transcript expression compared to untransfected U 87 glioma cells (Figure 1). By contrast, when the U87 cells were transfected with the NFIA- shRNAi mutant that contains five point mutations. In comparison with the mutant, the NFIA-RNAi demonstrated only a 75% reduction in transcript expression.
[0102] The data demonstrate that NFIA shRNA constructs can be used to reduce NFIA transcription in glioma cells.
[0103] The following example describes experiments to determine whether reduced levels of NFIA caused by NFIA-RNAi lead to an inhibition of growth in the U87 cell lines.
EXAMPLE 2 -NFlA SMALL HAIRPIN RNA REDUCES GROWTH OF GLIOMA CELL
LINES
[0104] To test the effects of NFIA antisense compositions on the growth of gliomas in vitro, theU87 cell line was transduced with the lentiviral NFIA-shRNAi viral supernatant, (described in Example 1) lentiviral NFIA-shRNAi-mutant viral supernatant (described in Example 1), or was left untreated.
[0105] As shown in Figure 2, transduction of gliomal cells with NFIA-shRNAi led to a dramatic reduction in cell growth compared to untreated U87 cells, and U87 cells transduced with the mutant-shRNAi lentivirus.
[0106] These data demonstrate that NFIA antisense inhibitors reduce growth of NFIA in gliomal cells, and demonstrate that NFlA antisense inhibitors are useful for the treatment of glioma.
[0107] The following example describes experiments to confirm whether NFIA plays an active role in tumorigenesis.
EXAMPLE 3 -NFlA SMALL HAIRPIN RNA REDUCES GLIOMAL TUMOR GROWTH
RATE IN VIVO
[0108] To test whether NFlA antisense inhibitors reduce the growth of gliomal tumors, U87 gliomal cells were transduced with the lentiviral NFIA-shRNAi viral supernatant, lentiviral NFIA-shRNAi-mutant viral supernatant, or a lentilviral NFIA cDNA viral supernatant, as described in Example 1. Approximately 2.5x105 cells were injected intracranially into mice, and tumor formation and growth was monitored using MRI.
[0109] The results of the MRI are shown in Figures 3 and 4. As shown in Figure 4A, over-expression of NFIA led to an accelerated rate of tumor growth, compared to tumor growth in mice injected with cells transduced with the lentiviral vector alone. Figure 4B. By contrast, knockdown of NFIA expression in U87 cells with the shRNAi NFIA inhibitor led to a dramatic reduction in tumor size. Figure 4D> Figure 3. The reduction was not as pronounced in mice injected with U87 cells transduced with the mutant shRNAi NFIA lentiviral supernatant. Figure 4C.
[0110] The data demonstrate that NFIA-RNAi NFIA inhibitor results in a dramatic reduction in the size and frequency of gliomal tumors.
[0111] The following example describes experiments to confirm that NFIA function is necessary for glioma formation, using a primary mouse neurosphere model of glioma.
EXAMPLE 4 -REDUCTION OF GLIOMAL TUMOR SIZE IN PRIMARY MOUSE NEUROSPHERE MODEL OF GLIOMA
[0112] In order to determine whether NFlA function is necessary for glioma formation in a primary mouse neurosphere model, transgenic NFIA mice (described in Neves et al Proc. Nat. Acad. Sci. USA (1999) 96(21): 11946-11951) was used to generate NFIA +/- and NFIA -/- neurospheres from E 11.5 telencephelon. Neurospheres were transduced with retroviruses expressing SV40 Large T antigen and constitutively activated EGFR-vIO. Expression of SV40 Large T antigen and EGFR-vIIl were validated via immunocytochemistry (not shown). NFIA +/-; SV40:EGFR-vIII and NFIA -;SV40;EGFR- vlll neurospheres were injected subcutaneously into the necks of SCID mice. Six weeks post-injection, tumors were harvested and analyzed. The results are presented in Table 1.
TABLE 1
Figure imgf000034_0001
[0113] The data demonstrate that NFIA function is necessary for glioma formation in a primary mouse neurposphere model. As such, the data confirm that NFIA is a useful therapeutic target for glioma. EXAMPLE 5 -REDUCTION OF GLIOMAL TUMOR SIZE
[0114] This example illustrates the reduction of the size of a gliomal tumor in a subject.
[0115] A subject is identified as having, or at risk of developing a glioma such as an astrocytoma using routine diagnostic methods. The subject is administered a daily dose of an NFIA inhibitor, such that the NFIA inhibitor contacts the glioma (e.g., by injection of the NFlA inhibitor into the glioma). The daily dose of the NFIA inhibitor can, in some cases, range from about 0.01 μg/kg to about 1 mg/kg of subject body weight or more per day, depending on the factors mentioned above. Preferably the dose of the NFIA inhibitor ranges from about 10 μg/kg/day to about 100 μg/kg/day. The appropriate dosage and treatment regimen can be readily determined by the skilled artisan based on a number of factors, including but not limited to the nature of the NFIA inhibitor, and the subject's disease state (e.g., the size and/or stage of the glioma), or condition. Levels of NFIA within the glioma can be determined before and after administration of the NFIA inhibitor. The glioma tumor size can be determined before and after administration of the NFIA inhibitor. EXAMPLE 6 -TREATMENT OF ASTROCYTOMA
[0116] This example illustrates the treatment of an astrocytoma in a subject.
[0117] A subject is identified as having, or at risk of developing an astrocytoma using routine diagnostic methods. The subject is administered a daily dose of an NFIA inhibitor, such that the NFlA inhibitor contacts the astrocytoma (e.g., by injection of the NFIA inhibitor into the astrocytoma). The daily dose of the NFIA inhibitor can, in some cases, range from about 0.01 μg/kg to about 1 mg/kg of subject body weight or more per day, depending on the factors mentioned above. Preferably the dose of the NFlA inhibitor ranges from about 10 μg/kg/day to about 100 μg/kg/day. The appropriate dosage and treatment regimen can be readily determined by the skilled artisan based on a number of factors, including but not limited to the nature of the NFIA inhibitor, and the subject's disease state (e.g., the size and/or stage of the tumor), or condition. Levels of NFIA within the astrocytoma can be determined before and after administration of the NFIA inhibitor. The astrocytoma tumor size can be determined before and after administration of the NFIA inhibitor. [0118] All patents and publications are herein incorporated by reference in their entireties to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference.
[0119] The invention illustratively described herein suitably can be practiced in the absence of nay element or elements, limitation or limitations that is not specifically disclosed herein. The terms and expression which have been employed are used as terms of description and not of limitation, and there is no intention that the use of such terms and expressions indicates the exclusion of equivalents of the features shown and described, or portions thereof. It is recognized that various modifications are possible within the scope of the invention disclosed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention.

Claims

WHAT IS CLAIMED IS:
1. Use of a therapeutically effective amount of an NFIA inhibitor in the manufacture of a medicament for reducing the size of a glioma.
2. The use of Claim I3 wherein said glioma is an astrocytoma.
3. The use of Claim 1 , wherein said NFIA inhibitor comprises an NFIA antisense polynucleotide.
4. The use of Claim 3, wherein said NFIA inhibitor comprises an NFIA short hairpin RNA (shRNA).
5. The use of Claim 4, wherein said shRNA targets human NFIA.
6. The use of Claim 5, wherein the NFIA shRNA comprises the sequence of SEQ ID NO: 1.
7. The use of Claim 3, wherein said NFlA inhibitor comprises a viral expression vector that comprises said NFIA antisense polynucleotide.
8. The use of Claim I3 wherein said medicament is formulated for injection into said glioma.
9. The use of Claim 1, wherein said NFIA inhibitor is an antibody that specifically binds to NFIA.
10. Use of a therapeutically effective amount of an NFIA inhibitor in the manufacture of medicament for treating a glioma in a subject in need thereof.
11. The use of Claim 10, wherein said glioma is an astrocytoma.
12. The use of Claim 10, wherein said NFIA inhibitor is an NFlA antisense polynucleotide.
13. The use of Claim 12, wherein said NFIA antisense oligonucleotide comprises an NFIA short hairpin RNA (shRNA).
14. The use of Claim 13, wherein the shRNA targets human NFIA.
15. The use of Claim 14, wherein said shRNA comprises SEQ ID NO: 1.
16. The use of Claim 12, wherein the NFIA inhibitor comprises a viral expression vector that comprises said NFIA antisense polynucleotide.
17. The use of Claim 10, wherein the medicament is formulated for injection into said glioma.
18. The use of Claim 10, wherein said NFIA inhibitor is an antibody that specifically binds to NFIA.
19. Use of a therapeutically effective amount of an NFlA inhibitor in the manufacture of a medicament for reducing the amount of functional NFIA in a glioma.
20. The use of Claim 19, wherein said NFIA inhibitor is an NFIA antisense polynucleotide.
21. The use of Claim 20, wherein said NFIA antisense oligonucleotide comprises an NFIA short hairpin RNA (shRNA).
22. The use of Claim 21, wherein the shRNA targets human NFIA.
23. The use of Claim 22, wherein said shRNA comprises SEQ ID NO: 1.
24. The use of Claim 20, wherein the NFIA inhibitor comprises a viral expression vector that comprises said NFIA antisense polynucleotide.
25. The use of Claim 10, wherein said NFIA inhibitor is an antibody that specifically binds to NFIA.
26. An antisense polynucleotide comprising an isolated polynucleotide comprising a nucleic acid that is at least 90% identical to SEQ ID NO: 1.
PCT/US2008/077119 2007-09-21 2008-09-19 Nfia in glial fate determination, glioma therapy and astrocytoma treatment WO2009039442A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US99475307P 2007-09-21 2007-09-21
US60/994,753 2007-09-21

Publications (2)

Publication Number Publication Date
WO2009039442A1 true WO2009039442A1 (en) 2009-03-26
WO2009039442A9 WO2009039442A9 (en) 2010-11-18

Family

ID=40468405

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/077119 WO2009039442A1 (en) 2007-09-21 2008-09-19 Nfia in glial fate determination, glioma therapy and astrocytoma treatment

Country Status (2)

Country Link
US (1) US7951785B2 (en)
WO (1) WO2009039442A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010125107A1 (en) * 2009-04-28 2010-11-04 Life & Brain Gmbh Microglial precursor cells for the treatment of malignant neoplasms of the central nervous system

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016154344A1 (en) 2015-03-24 2016-09-29 The Regents Of The University Of California Adeno-associated virus variants and methods of use thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001055162A1 (en) * 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
WO2005005601A2 (en) * 2003-06-09 2005-01-20 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US20070213286A1 (en) * 2005-09-12 2007-09-13 The Trustees Of Columbia University In The City Of New York Inhibitors of dynein or lissencephaly 1, and methods of using same for treatment of neuronal disorders

Family Cites Families (161)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US4476301A (en) 1982-04-29 1984-10-09 Centre National De La Recherche Scientifique Oligonucleotides, a process for preparing the same and their application as mediators of the action of interferon
JPS5927900A (en) 1982-08-09 1984-02-14 Wakunaga Seiyaku Kk Oligonucleotide derivative and its preparation
FR2540122B1 (en) 1983-01-27 1985-11-29 Centre Nat Rech Scient NOVEL COMPOUNDS COMPRISING A SEQUENCE OF OLIGONUCLEOTIDE LINKED TO AN INTERCALATION AGENT, THEIR SYNTHESIS PROCESS AND THEIR APPLICATION
US4605735A (en) 1983-02-14 1986-08-12 Wakunaga Seiyaku Kabushiki Kaisha Oligonucleotide derivatives
US4948882A (en) 1983-02-22 1990-08-14 Syngene, Inc. Single-stranded labelled oligonucleotides, reactive monomers and methods of synthesis
US4824941A (en) 1983-03-10 1989-04-25 Julian Gordon Specific antibody to the native form of 2'5'-oligonucleotides, the method of preparation and the use as reagents in immunoassays or for binding 2'5'-oligonucleotides in biological systems
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4587044A (en) 1983-09-01 1986-05-06 The Johns Hopkins University Linkage of proteins to nucleic acids
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
US5118802A (en) 1983-12-20 1992-06-02 California Institute Of Technology DNA-reporter conjugates linked via the 2' or 5'-primary amino group of the 5'-terminal nucleoside
US5550111A (en) 1984-07-11 1996-08-27 Temple University-Of The Commonwealth System Of Higher Education Dual action 2',5'-oligoadenylate antiviral derivatives and uses thereof
FR2567892B1 (en) 1984-07-19 1989-02-17 Centre Nat Rech Scient NOVEL OLIGONUCLEOTIDES, THEIR PREPARATION PROCESS AND THEIR APPLICATIONS AS MEDIATORS IN DEVELOPING THE EFFECTS OF INTERFERONS
JPS6147500A (en) 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
US5258506A (en) 1984-10-16 1993-11-02 Chiron Corporation Photolabile reagents for incorporation into oligonucleotide chains
US5430136A (en) 1984-10-16 1995-07-04 Chiron Corporation Oligonucleotides having selectably cleavable and/or abasic sites
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
US4828979A (en) 1984-11-08 1989-05-09 Life Technologies, Inc. Nucleotide analogs for nucleic acid labeling and detection
JPS61134325A (en) 1984-12-04 1986-06-21 Teijin Ltd Expression of hybrid antibody gene
FR2575751B1 (en) 1985-01-08 1987-04-03 Pasteur Institut NOVEL ADENOSINE DERIVATIVE NUCLEOSIDES, THEIR PREPARATION AND THEIR BIOLOGICAL APPLICATIONS
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5405938A (en) 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5166315A (en) 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US4762779A (en) 1985-06-13 1988-08-09 Amgen Inc. Compositions and methods for functionalizing nucleic acids
AU6131086A (en) 1985-07-05 1987-01-30 Whitehead Institute For Biomedical Research Epithelial cells expressing foreign genetic material
US4980286A (en) 1985-07-05 1990-12-25 Whitehead Institute For Biomedical Research In vivo introduction and expression of foreign genetic material in epithelial cells
US5317098A (en) 1986-03-17 1994-05-31 Hiroaki Shizuya Non-radioisotope tagging of fragments
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
JPS638396A (en) 1986-06-30 1988-01-14 Wakunaga Pharmaceut Co Ltd Poly-labeled oligonucleotide derivative
DE3788914T2 (en) 1986-09-08 1994-08-25 Ajinomoto Kk Compounds for cleaving RNA at a specific position, oligomers used in the preparation of these compounds and starting materials for the synthesis of these oligomers.
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5166320A (en) 1987-04-22 1992-11-24 University Of Connecticut Carrier system and method for the introduction of genes into mammalian cells
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
ATE113059T1 (en) 1987-06-24 1994-11-15 Florey Howard Inst NUCLEOSIDE DERIVATIVES.
US5585481A (en) 1987-09-21 1996-12-17 Gen-Probe Incorporated Linking reagents for nucleotide probes
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
US5525465A (en) 1987-10-28 1996-06-11 Howard Florey Institute Of Experimental Physiology And Medicine Oligonucleotide-polyamide conjugates and methods of production and applications of the same
DE3738460A1 (en) 1987-11-12 1989-05-24 Max Planck Gesellschaft MODIFIED OLIGONUCLEOTIDS
US5403711A (en) 1987-11-30 1995-04-04 University Of Iowa Research Foundation Nucleic acid hybridization and amplification method for detection of specific sequences in which a complementary labeled nucleic acid probe is cleaved
JP3019994B2 (en) 1987-11-30 2000-03-15 ユニバーシティ オブ アイオワ リサーチ ファウンデーション Novel oligodeoxynucleotide, method of blocking expression of target gene using the same, and novel oligodeoxynucleotide and method of inhibiting expression of target gene using the same
US5082830A (en) 1988-02-26 1992-01-21 Enzo Biochem, Inc. End labeled nucleotide probe
EP0406309A4 (en) 1988-03-25 1992-08-19 The University Of Virginia Alumni Patents Foundation Oligonucleotide n-alkylphosphoramidates
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5109124A (en) 1988-06-01 1992-04-28 Biogen, Inc. Nucleic acid probe linked to a label having a terminal cysteine
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5262536A (en) 1988-09-15 1993-11-16 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-tagged oligonucleotides
US5194599A (en) 1988-09-23 1993-03-16 Gilead Sciences, Inc. Hydrogen phosphonodithioate compositions
US5512439A (en) 1988-11-21 1996-04-30 Dynal As Oligonucleotide-linked magnetic particles and uses thereof
US5457183A (en) 1989-03-06 1995-10-10 Board Of Regents, The University Of Texas System Hydroxylated texaphyrins
US5599923A (en) 1989-03-06 1997-02-04 Board Of Regents, University Of Tx Texaphyrin metal complexes having improved functionalization
US5391723A (en) 1989-05-31 1995-02-21 Neorx Corporation Oligonucleotide conjugates
US5256775A (en) 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US4958013A (en) 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5451463A (en) 1989-08-28 1995-09-19 Clontech Laboratories, Inc. Non-nucleoside 1,3-diol reagents for labeling synthetic oligonucleotides
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5254469A (en) 1989-09-12 1993-10-19 Eastman Kodak Company Oligonucleotide-enzyme conjugate that can be used as a probe in hybridization assays and polymerase chain reaction procedures
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
US5721218A (en) 1989-10-23 1998-02-24 Gilead Sciences, Inc. Oligonucleotides with inverted polarity
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
AU658562B2 (en) 1989-10-24 1995-04-27 Isis Pharmaceuticals, Inc. 2' modified oligonucleotides
US5292873A (en) 1989-11-29 1994-03-08 The Research Foundation Of State University Of New York Nucleic acids labeled with naphthoquinone probe
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5486603A (en) 1990-01-08 1996-01-23 Gilead Sciences, Inc. Oligonucleotide having enhanced binding affinity
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5578718A (en) 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5623065A (en) 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5149797A (en) 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
US5220007A (en) 1990-02-15 1993-06-15 The Worcester Foundation For Experimental Biology Method of site-specific alteration of RNA and production of encoded polypeptides
US5214136A (en) 1990-02-20 1993-05-25 Gilead Sciences, Inc. Anthraquinone-derivatives oligonucleotides
AU7579991A (en) 1990-02-20 1991-09-18 Gilead Sciences, Inc. Pseudonucleosides and pseudonucleotides and their polymers
US5321131A (en) 1990-03-08 1994-06-14 Hybridon, Inc. Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
GB9009980D0 (en) 1990-05-03 1990-06-27 Amersham Int Plc Phosphoramidite derivatives,their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
EP0745689A3 (en) 1990-05-11 1996-12-11 Microprobe Corporation A dipstick for a nucleic acid hybridization assay
US5688941A (en) 1990-07-27 1997-11-18 Isis Pharmaceuticals, Inc. Methods of making conjugated 4' desmethyl nucleoside analog compounds
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
BR9106702A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Inc ANALOG OF OLIGONUCLEOTIDEOS AND PROCESSES TO MODULATE THE PRODUCTION OF A PROTEIN BY AN ORGANISM AND TO TREAT AN ORGANISM
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5245022A (en) 1990-08-03 1993-09-14 Sterling Drug, Inc. Exonuclease resistant terminally substituted oligonucleotides
CA2088673A1 (en) 1990-08-03 1992-02-04 Alexander L. Weis Compounds and methods for inhibiting gene expression
US5177196A (en) 1990-08-16 1993-01-05 Microprobe Corporation Oligo (α-arabinofuranosyl nucleotides) and α-arabinofuranosyl precursors thereof
US5512667A (en) 1990-08-28 1996-04-30 Reed; Michael W. Trifunctional intermediates for preparing 3'-tailed oligonucleotides
US5214134A (en) 1990-09-12 1993-05-25 Sterling Winthrop Inc. Process of linking nucleosides with a siloxane bridge
US5561225A (en) 1990-09-19 1996-10-01 Southern Research Institute Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
EP0549686A4 (en) 1990-09-20 1995-01-18 Gilead Sciences Inc Modified internucleoside linkages
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
CA2095212A1 (en) 1990-11-08 1992-05-09 Sudhir Agrawal Incorporation of multiple reporter groups on synthetic oligonucleotides
US5672697A (en) 1991-02-08 1997-09-30 Gilead Sciences, Inc. Nucleoside 5'-methylene phosphonates
DE69230142T2 (en) 1991-05-15 2000-03-09 Cambridge Antibody Tech METHOD FOR PRODUCING SPECIFIC BINDING PAIRS
US5539082A (en) 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5714331A (en) 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5719262A (en) 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
US5371241A (en) 1991-07-19 1994-12-06 Pharmacia P-L Biochemicals Inc. Fluorescein labelled phosphoramidites
US5571799A (en) 1991-08-12 1996-11-05 Basco, Ltd. (2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
EP0538194B1 (en) 1991-10-17 1997-06-04 Novartis AG Bicyclic nucleosides, oligonucleotides, their method of preparation and intermediates therein
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
AU3222793A (en) 1991-11-26 1993-06-28 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
TW393513B (en) 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5792608A (en) 1991-12-12 1998-08-11 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
US5700922A (en) 1991-12-24 1997-12-23 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
US5565552A (en) 1992-01-21 1996-10-15 Pharmacyclics, Inc. Method of expanded porphyrin-oligonucleotide conjugate synthesis
US5595726A (en) 1992-01-21 1997-01-21 Pharmacyclics, Inc. Chromophore probe for detection of nucleic acid
FR2687679B1 (en) 1992-02-05 1994-10-28 Centre Nat Rech Scient OLIGOTHIONUCLEOTIDES.
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5633360A (en) 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
EP0577558A2 (en) 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclic nucleosides having bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
US5272250A (en) 1992-07-10 1993-12-21 Spielvogel Bernard F Boronated phosphoramidate compounds
US5652355A (en) 1992-07-23 1997-07-29 Worcester Foundation For Experimental Biology Hybrid oligonucleotide phosphorothioates
US5574142A (en) 1992-12-15 1996-11-12 Microprobe Corporation Peptide linkers for improved oligonucleotide delivery
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
GB9304618D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
DK0691968T3 (en) 1993-03-30 1998-02-23 Sanofi Sa Acyclic nucleoside analogs and oligonucleotide sequences containing these
JPH08508491A (en) 1993-03-31 1996-09-10 スターリング ウインスロップ インコーポレイティド Oligonucleotides with phosphodiester bonds replaced by amide bonds
DE4311944A1 (en) 1993-04-10 1994-10-13 Degussa Coated sodium percarbonate particles, process for their preparation and detergent, cleaning and bleaching compositions containing them
FR2705099B1 (en) 1993-05-12 1995-08-04 Centre Nat Rech Scient Phosphorothioate triester oligonucleotides and process for their preparation.
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
US5446137B1 (en) 1993-12-09 1998-10-06 Behringwerke Ag Oligonucleotides containing 4'-substituted nucleotides
ATE196311T1 (en) 1993-12-09 2000-09-15 Univ Jefferson COMPOUNDS AND METHODS FOR SITE-SPECIFIC MUTATION IN EUKARYOTIC CELLS
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5627053A (en) 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5646269A (en) 1994-04-28 1997-07-08 Gilead Sciences, Inc. Method for oligonucleotide analog synthesis
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
US5597696A (en) 1994-07-18 1997-01-28 Becton Dickinson And Company Covalent cyanine dye oligonucleotide conjugates
US5580731A (en) 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5597909A (en) 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5792747A (en) 1995-01-24 1998-08-11 The Administrators Of The Tulane Educational Fund Highly potent agonists of growth hormone releasing hormone
US5652356A (en) 1995-08-17 1997-07-29 Hybridon, Inc. Inverted chimeric and hybrid oligonucleotides
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US7195916B2 (en) 2001-09-13 2007-03-27 California Institute Of Technology Method for expression of small antiviral RNA molecules within a cell

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001055162A1 (en) * 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
WO2005005601A2 (en) * 2003-06-09 2005-01-20 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US20070213286A1 (en) * 2005-09-12 2007-09-13 The Trustees Of Columbia University In The City Of New York Inhibitors of dynein or lissencephaly 1, and methods of using same for treatment of neuronal disorders

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010125107A1 (en) * 2009-04-28 2010-11-04 Life & Brain Gmbh Microglial precursor cells for the treatment of malignant neoplasms of the central nervous system

Also Published As

Publication number Publication date
US20090087434A1 (en) 2009-04-02
US7951785B2 (en) 2011-05-31
WO2009039442A9 (en) 2010-11-18

Similar Documents

Publication Publication Date Title
US11279933B2 (en) Antisense oligonucleotides for the treatment of leber congenital amaurosis
AU2017210563B2 (en) Compositions and methods for inhibiting gene expression of hepatitis b virus
ES2908962T3 (en) Antisense oligonucleotides useful in the treatment of Pompe disease
US20090270486A1 (en) Human rnase iii and compositions and uses thereof
CA3099522A1 (en) Gapmers and methods of using the same for treatment of muscular dystrophy
CN110997916A (en) Antisense oligonucleotides for treating Stargardt&#39;s disease
Graham et al. Towards a therapeutic inhibition of dystrophin exon 23 splicing in mdx mouse muscle induced by antisense oligoribonucleotides (splicomers): target sequence optimisation using oligonucleotide arrays
JP2011527893A (en) Compositions and methods for inhibiting expression of TGF-β receptor gene
US20210322526A1 (en) Enzymatic replacement therapy and antisense therapy for pompe disease
US7951785B2 (en) NFIA in glial fate determination, glioma therapy and astrocytoma treatment
US9205102B2 (en) Method for treatment of castration-resistant prostate cancer
US20020164601A1 (en) Human RNase III and compositions and uses thereof
EP3790893A1 (en) Methods for improving leptin sensitivity for the treatment of obesity and diabetes
US20050164234A1 (en) Methods of using mammalian RNase H and compositions thereof
WO2018102736A1 (en) Methods for the treatment of cancer
US20230287427A1 (en) Inhibition of lncExACT1 to Treat Heart Disease
JP2003521923A (en) Selective killing of cells by activation of double-stranded RNA-dependent protein kinase-PKR

Legal Events

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

Ref document number: 08832758

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08832758

Country of ref document: EP

Kind code of ref document: A1