US20070219152A1 - Inhibition of Syk kinase expression - Google Patents

Inhibition of Syk kinase expression Download PDF

Info

Publication number
US20070219152A1
US20070219152A1 US11/639,243 US63924306A US2007219152A1 US 20070219152 A1 US20070219152 A1 US 20070219152A1 US 63924306 A US63924306 A US 63924306A US 2007219152 A1 US2007219152 A1 US 2007219152A1
Authority
US
United States
Prior art keywords
sirna
cells
syk kinase
expression
syk
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/639,243
Inventor
Alan Schreiber
Zena Indik
Moo-Kyung Kim
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Pennsylvania Penn
Original Assignee
University of Pennsylvania Penn
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Pennsylvania Penn filed Critical University of Pennsylvania Penn
Priority to US11/639,243 priority Critical patent/US20070219152A1/en
Publication of US20070219152A1 publication Critical patent/US20070219152A1/en
Priority to US13/067,035 priority patent/US20120093913A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/10Protein-tyrosine kinases (2.7.10)
    • C12Y207/10001Receptor protein-tyrosine kinase (2.7.10.1)
    • 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.

Definitions

  • the present invention relates, in general, to Syk kinase and, in particular, to a method of inhibiting Syk kinase expression using small interfering RNA (siRNA).
  • siRNA small interfering RNA
  • Double stranded RNA has been shown to be a powerful agent for interfering with gene expression in a number of organisms, including C. elegans and Drosophila, as well as plants (Bernstein et al, RNA 7:1509-2151 (2001), McManus et al, Nat. Rev. Genet. 3:737-747 (2992), Hutvagner et al, Curr. Opin. Genet. Dev. 12:225-232 (2002), Zamore, Nat. Struct. Biol. 8:746-750 (2001) Tuschl et al, Genes Dev. 13:3191-3197 (1999)).
  • siRNAs can function well as gene silencing agents in mammalian cells (Elbashir et al, Nature 411:494-498 (2001), Elbashir et al, Genes Dev. 15:188-200 (2001), Paddison et al, Genes Dev. 16:948-958 (2002), Wianny et al, Nat. Cell Biol. 2:70-75 (2000)).
  • RNAi involves a multi-step process. Double stranded RNAs are cleaved by the endonuclease Dicer to generate 21-23 nucleotide fragments (siRNA). The siRNA duplex is resolved into 2 single stranded RNAs, one strand being incorporated into a protein-containing complex where it functions as guide RNA to direct cleavage of the target RNA (Schwarz et al, Mol. Cell. 10:537-548 (2002), Zamore et al, Cell 101:25-33 (2000)), thus silencing a specific genetic message (see also Zeng et al, Proc. Natl. Acad. Sci. 100:9779 (2003)).
  • Anti-sense DNA has also been widely used to inhibit gene expression (Roth et al, Annu. Rev. Biomed. Eng. 1:265-297 (1999)). Once inside the cell, anti-sense oligonucleotides (ASO) recognize, then bind tightly to complementary mRNA, thus preventing the mRNA from interacting with the protein translation machinery of the cell.
  • ASO anti-sense oligonucleotides
  • siRNA is more potent and reliable than ASO as an inhibitor of gene expression.
  • the present invention results from studies designed to test the efficacy of siRNA as an inhibitor of Syk kinase expression.
  • the present invention relates generally to Syk kinase.
  • the invention relates to a method of inhibiting Syk kinase expression using small interfering RNA (siRNA) and to therapeutic strategies based on such a method.
  • siRNA small interfering RNA
  • FIG. 1 The sense strand of each Syk kinase siRNA is the same sequence as the target sequence with the exception of the initial template adenine dimer and terminal overhang thymidine dimer.
  • the antisense strand of the siRNA is the reverse complement of the target sequence.
  • FIG. 2 Expression of Syk kinase in RBL-2H3 cells transfected with siRNA targeted to Syk kinase mRNA.
  • RBL-2H3 cells were transfected with siRNA-1 (Lane 2), siRNA-2 (lane 3) or lipofectamine transfection control (Lane 1). Proteins in cell lysates were separated by SDS-PAGE and transferred to nitrocellulose. Top panel, Syk kinase immunoblot; bottom panel, actin immunoblot.
  • FIG. 3 Expression of Syk kinase in human monocytes transfected with siRNA targeted to Syk kinase mRNA.
  • Monocytes were treated with siRNA (Lane 2) or lipofectamine transfection control (Lane 1). Proteins in cell lysates were separated by SDS-PAGE, transferred to nitrocellulose and immunoblotted with anti-Syk kinase antibody.
  • FIG. 4 Western blot analysis: Syk protein expression in HS-24 cells.
  • FIGS. 5A and 5B HS-24 cells, following siRNA treatment, were lysed and equal amounts of total protein in HS-24 cell lysates were resolved by 10% SDS gel electrophoresis, and analyzed by Western blot using monoclonal antibody to Syk or actin. Lane 1—no treatment, lane 2—siRNA-1 (control) 24 h treatment, lane 3—siRNA-1 48 h treatment, lane 4—siRNA-2 24 h treatment, lane 5—siRNA-2 48 h treatment. ( FIG. 5B ) RNA was isolated and RT-PCR was performed for Syk and ⁇ -actin. Lane 1—no treatment, lane 2—siRNA-1 (control) 48 h treatment, lane 3—siRNA-2 48 h treatment.
  • FIGS. 6A and 6B HS-24 cells plated on either polylysine coated plates (non stimulated, resting) or fibronectin coated plates (stimulated) were treated with siRNA-2, or siRNA-1 (control), or piceatannol. Cells were treated with 10 ng/ml of TNF during overnight culture.
  • FIG. 6A Following siRNA (48 h) or piceatannol (16 h) treatment, cells were removed, immunostained with anti-CD54 (ICAM-1) and analysed by flow cytometry.
  • FIG. 6B Cell culture supernatants were analysed for IL-6 release using an IL-6 ELISA kit.
  • FIGS. 7A and 7B Effect of siRNA targeted to Syk kinase delivered via aerosol on total cell numbers in bronchoalveolar lavage (BAL) fluid of ovalbumin (OA) sensitized and challenged Brown Norway Rats after three treatments.
  • BAL bronchoalveolar lavage
  • OA ovalbumin
  • FIG. 7A provides data as bar graphs
  • FIG. 7B shows individual data points (individual animals).
  • FIGS. 8A-8D Effect of siRNA targeted to Syk kinase delivered via aerosol on numbers of macrophages, neutrophils, lymphocytes and eosinophils in BAL fluid of OA sensitized and challenged Brown Norway rats after three treatments.
  • FIG. 8A provides data as bar graphs
  • FIG. 8B shows individual data points (individual animals) for macrophage numbers
  • FIG. 8C shows individual data points for neutrophil numbers
  • FIG. 8D shows individual data points for eosinophil numbers.
  • the present invention relates to RNA molecules that target Syk kinase mRNA.
  • the invention relates to RNA molecules about 19, 20 or 21 to about 23 nucleotides in length that direct cleavage and/or degradation of Syk kinase mRNA.
  • the present invention relates to the use of siRNA molecules, double stranded RNA molecules typically comprising two 20-23 nucleotide (nt) strands.
  • siRNAs suitable for use in the invention can be produced using any of a variety of approaches.
  • the siRNA can be prepared in vitro and then introduced directly into cells (for example, by transfection). Alternatively, intracellular expression can be effected by transfecting into cells constructs (e.g., DNA-based vectors or cassettes) that express siRNA within cells.
  • siRNA suitable for use in the invention can be prepared, for example, via chemical synthesis, in vitro transcription, enzymatic digestion of a longer dsRNA using an RNase III enzyme such as Dicer or RNase III, expression in cells from an siRNA expression plasmid or viral vector, or expression in cells from a PCR-derived siRNA expression cassette.
  • RNase III enzyme such as Dicer or RNase III
  • RNA of the invention Various approaches are available to enhance stability of RNA of the invention, (see, for example, U.S. Application Nos. 20020086356, 20020177570 and 20020055162, and U.S. Pat. Nos. 6,197,944, 6,590,093, 6,399,307, 6,057,134, 5,939,262, and 5,256,555, and references cited therein).
  • siRNA suitable for use in the invention can be prepared chemically.
  • 2′ hydroxyls are protected during the synthetic process against degradation using, for example, acid labile orthoester protecting groups (see Scaringe et al, J. Am. Chem. Soc. 120:11820 (1998) and www.dharmacon.com (e.g., the ACE technology described therein)).
  • the RNA oligomers can be simultaneously 2′ deprotected and annealed prior to use.
  • At least one strand of the double stranded molecule can have a 3′ overhang from about 1 to about 6 nucleotides (e.g., pyrimidine and/or purine nucleotides) in length.
  • the 3′ overhang is from about 1 to about 5 nucleotides (e.g., thymidines or uridines), more preferably from about 1 to about 4 nucleotides and most preferably 2 or 3 nucleotides in length.
  • each strand has an overhang.
  • the length of the overhangs can be the same or different for each strand. Typically, both strands have overhangs of the same length.
  • the RNA of the present invention comprises 21 or 22 nucleotide strands that are paired and that have overhangs of from about 1 to about 3, particularly, about 2, nucleotides on the 3′ ends of both of the RNA strands.
  • siRNAs suitable for use in the invention can be prepared by enzymatic digestion of a longer dsRNA using an RNase III type enzyme (e.g., Dicer).
  • Dicer an RNase III type enzyme
  • a commercially available Dicer siRNA generation kit can be used that permits generation of large numbers of siRNAs from full length target genes (Gene Therapy Systems, Inc, MV062603).
  • SiRNA can be produced from target DNA and T7 RNA polymerase promoter sequences using PCR based cloning. Following RNA transcription from the target sequence, recombinant Dicer can cleave the transcribed RNAi into 22 bp siRNAs.
  • siRNA molecules suitable for use in the present invention can also be recombinantly produced using methods known in the art. (See references and web sites cited above.) Recombinant technology permits in vivo transcription of siRNAs in mammalian cells.
  • vectors can be used that contain, for example, RNA polymerase III or U6 promoter sequences.
  • Such vectors can be used as expression vectors or as shuttle vectors in conjunction with viral systems (e.g., adenoviral or retroviral systems) to introduce siRNA into mammalian cells.
  • Vectors can be engineered to express sense and anti-sense strands of siRNAs that anneal in vivo to produce functional siRNAs.
  • hairpin RNA can be expressed by inserting into a vector the sense strand (e.g., about 20 nt) of the target, followed by a short spacer (e.g., about 4 to about 10 nt), then the antisense strand of the target (e.g., about 20 nt) and, for example, about 5-6 T's as transcription terminator.
  • the resulting RNA transcript folds back to form a stem-loop structure comprising, for example, about a 20 bp stem and about a 10 nt loop with 2-3 U's at the 3′ end.
  • Constructs suitable for use in effecting in vivo production can be readily designed by one skilled in the art and will vary, for example, with the cell/tissue target and the effect sought.
  • dsRNA can be used in the methods of the present invention provided it has sufficient homology to the targeted Syk kinase mRNA.
  • SiRNA duplexes can be designed, for example, by searching Syk kinase cDNA for the target motif “AA(N) 19 ”, wherein N is any nucleotide, motifs with approximately 30% to 70% G/C content being preferred, those of about 50% G/C content being more preferred.
  • the sense strand of the siRNA duplex can correspond to nucleotides 3 to 21 of the selected AA(N) 19 motif.
  • the antisense strand of the siRNA duplex can have a sequence complementary to nucleotides 1 to 21 of the selected AA(N) 19 motif. Further design details are provided at http://www.mpibpc.gwdg.de/ en/100/105/sirna.html.
  • target sequences include sequences unique to Syk kinase mRNA.
  • target sequences can be selected from sequences between the two SH2 domains of Syk kinase or between the second SH2 domain and the kinase domain. Certain specific DNA target sequences are described in the non-limiting Examples that follow.
  • Additional targets include, but are not limited to, the following: Identified homologies of *Sequence % GC 16-18/19 nucleotides AATATGTGAAGCAGACATGGA 42 mitochondrial ribosomal prot15 AATCAAATCATACTCCTTCCC 42 AAGAGAGTACTGTGTCATTCA 42 AAGGAAAACCTCATCAGGGAA 47 inositolhexaphosphate kinase, ⁇ globin on Chr11 AATCATACTCCTTCCCAAAGC 47 AATTTTGGAGGCCGTCCACAA 53 oxytokinase AAGACTGGGCCCTTTGAGGAT 58 AAGCACACATGGAACCTGCAG 58 histamine receptor H3, GTP binding protein AACTTCCAGGTTCCCATCCTG 58 AAGCCTGGCCACAGAAAGTCC 63 AAGCCCTACCCATGGACACAG 63 AACCTGCAGGGTCAGGCTCTG 68 AAGGGGTGCAGCCCAAGACTG 68 ⁇ glutamyl
  • siRNAs described herein can be used in a variety of ways.
  • the siRNA molecules can be used to target Syk kinase mRNA in a cell or organism.
  • the siRNA can be introduced into human cells or a human in order to mediate RNAi in the cells or in cells in the individual, so as to prevent or treat a disease or undesirable condition associated with Syk kinase expression (e.g., inflammation of the lungs, joints eyes or bladder).
  • a disease or undesirable condition associated with Syk kinase expression e.g., inflammation of the lungs, joints eyes or bladder.
  • the siRNA can also be used in the treatment of the immune destruction of blood cells, e.g., red blood cells in autoimmune hemolytic anemia and platelets in immune thrombocytopenic purpurea (ITP) (e.g., by targeting Syk kinase mRNA in macrophages and spleen and liver cells).
  • ITP immune thrombocytopenic purpurea
  • the Syk kinase gene is targeted and the corresponding mRNA (the transcriptional product of the targeted Syk kinase gene) is degraded by RNAi.
  • an siRNA-containing composition can be aerosolized and administered, for example, via inhalation. Administration to joints can be effected by injection of an siRNA-containing solution.
  • Administration to the eyes can be effected, for example, by injection or by application of drops comprising the siRNA in a carrier.
  • Administration to the bladder, etc. can be effected, for example, by washing or irrigating the target tissue with a composition containing the siRNA.
  • Administration to the skin can be via topical administration (e.g., as a liquid, cream or gel).
  • cells of an individual e.g., blood mononuclear cells, basophiles or mast cells
  • the cells to be treated can be obtained from the individual using known methods and the siRNAs that mediate degradation of the corresponding Syk kinase mRNA can be introduced into the cells, which can then be re-introduced into the individual.
  • the invention relates to the use of the above-described siRNAs to inhibit mediator (e.g., histamine) release from cells bearing an Fc ⁇ receptor, such as mast cells.
  • mediator e.g., histamine
  • histamine a mast cell mediator
  • siRNAs or constructs suitable for use in effecting intracellular production of siRNA of the invention can be administered systemically (e.g., via IV) or directly to the target tissue (e.g., via aerosol administration to the lung). Delivery can be effected using the techniques described herein (including liposome formulations). In addition to liposome formulations, polymer formulations can be used. Polyethylenimine (PEI) is an example of a suitable cationic polymer. Varying sizes of PEI can be used, including linear 22 kDa and branched 25 kDa PEI (other sizes, modified and unmodified, as well as biodegradable versions can be used).
  • PEI Polyethylenimine
  • Varying sizes of PEI can be used, including linear 22 kDa and branched 25 kDa PEI (other sizes, modified and unmodified, as well as biodegradable versions can be used).
  • Delivery can also be effected using, for example, non-toxic viral delivery systems (e.g., an adeno-associated viral delivery system). Optimum dosing will depend on the patient, the siRNA, the mode of administration, and the effect sought. Optimum conditions can be established by one skilled in the art without undue experimentation.
  • non-toxic viral delivery systems e.g., an adeno-associated viral delivery system.
  • Optimum dosing will depend on the patient, the siRNA, the mode of administration, and the effect sought. Optimum conditions can be established by one skilled in the art without undue experimentation.
  • Lipofectamine 2000 and Opti-Mem were purchased from Invitrogen (San Diego, Calif.).
  • Eagle's MEM (EMEM), FCS, penicillin, and streptomycin were purchased from Life Technologies (Grand Island, N.Y.).
  • Rabbit anti-murine Syk kinase polyclonal antibodies (Ab) and anti-actin Ab were purchased from Santa Cruz Biotechnology (Santa Cruz, Calif.), and F(ab′) 2 goat anti-rabbit Ab was supplied by The Jackson Laboratory (Bar Harbor, Me.).
  • Chemiluminescence reagent was purchased from DuPont NEN (Boston, Mass.).
  • Peripheral blood monocytes obtained from healthy volunteers at the University of Pennsylvania were isolated as previously described (Matsuda et al, Molec. Biol. of the Cell 7:1095-1106 (1996)). Briefly, heparinized blood was centrifuged on Ficoll-Hypaque (Lymphocyte Separation Medium; Organon Teknika, Durham, N.C.) and interface cells were resuspended in complete medium containing RPMI 1640 (GIBCO BRL, Grand Island, N.Y.), 10% heat inactivated-fetal calf serum (FCS) (Intergen, Purchase, N.Y.) and 2 mM L-glutamine. Cells were allowed to adhere at 37° C.
  • Ficoll-Hypaque Lymphocyte Separation Medium
  • FCS heat inactivated-fetal calf serum
  • Rat basophilic cells were grown in EMEM supplemented with 17% FBS, 100 U penicillin, 100 ⁇ g/ml streptomycin, and 4 mM glutamine at 37° C. in 5% CO 2 .
  • siRNAs for Syk kinase were prepared by Dharmacon Research Inc. (Lafayette, Colo.). In designing the siRNAs according to the guidelines provided by the manufacturer, potential siRNA targets (19 nucleotides immediately downstream of AA pairs) in human Syk kinase RNA were first identified. These sites were then scanned in the sequences of rat and mouse Syk kinase RNA in order to identify common Syk kinase RNA target sequences in these species. Two appropriate sites were identified, and two 21-mer RNAs, each consisting of 19 complementary nucleotides and 3′ terminal noncomplementary dimers of thymidine (Elbashir et al, Nature 411:494-498 (2001)), were constructed.
  • siRNA-1 Human, bp 296 to bp 316; Mouse and Rat, bp 307 to bp 327.
  • siRNA-2 Human, bp 364 to bp 382; Mouse and Rat, bp 375 to bp 393 sense 5′- ccucaucagggaauaucug dTdT 3′ antisense 3′-dTdT ggaguagucccuuauagac 5′ Transfections
  • siRNA was introduced into RBL-2H3 cells and into monocytes by transfection.
  • 5 ⁇ 10 4 RBL-2H3 cells or 1 ⁇ 10 5 monocytes were seeded into each well of a 24-well plate. Twenty-four hours later, the complete medium was replaced with 400 ⁇ l fresh medium lacking serum and antibiotic and siRNA/Lipofectamine 2000 complex was added to each well.
  • the siRNA/Lipofectamine 2000 complex was formed by adding 3 ⁇ l of siRNA duplex (20 ⁇ M) and 3 ⁇ l of Lipofectamine 2000 to 100 ⁇ l Opti-mem without serum or antibiotic according to the manufacturer's protocol.
  • the siRNA/Lipofectamine 2000 complex was formed by adding 3 ⁇ l of siRNA duplex (20 ⁇ M) and 1 ⁇ l of Lipofectamine 2000 to 100 ⁇ l Opti-mem without serum or antibiotic. Cells were incubated at 37° C. for 48 hours before examination of kinase protein expression by Western blotting.
  • Lysates were prepared by boiling cells in Laemmli sample buffer (2% SDS, 10% glycerol, 100 mM DTT, and 60 mM Tris (pH 6.8) for 5 minutes. Proteins in lysates were separated by SDS-PAGE (10% polyacrylamide) and transferred to a nitrocellulose membrane in sample buffer (25 mM Tris, 190 mM glycine, and 20% methanol). The nitrocellulose membrane was incubated overnight at 4° C. with 1 ⁇ g/ml rabbit anti-murine Syk kinsae polyclonal Ab before incubation with goat anti-rabbit HRP (1.5 h at room temperature).
  • Laemmli sample buffer 2% SDS, 10% glycerol, 100 mM DTT, and 60 mM Tris (pH 6.8) for 5 minutes. Proteins in lysates were separated by SDS-PAGE (10% polyacrylamide) and transferred to a nitrocellulose membrane in sample buffer (25 mM Tris, 190
  • Protein bands on the membrane were visualized with chemiluminescence reagent according to the manufacturer's protocol.
  • the anti-Syk kinase Ab was removed by incubating the membrane in a stripping buffer containing 100 mM 2-ME, 2% SDS, and 62.5 mM Tris-HCL (pH 6.7) for 30 minutes at 50° C. with occasional agitation.
  • the membrane was then reprobed with anti-actin Ab and bands on the membrane were visualized with chemiluminescence reagent. Protein levels of Syk kinase were quantified by densitometry (Personal Densitometer, Molecular Dynamics).
  • Rat basophilic cells RBL-2H3 cells
  • human monocytes were transfected with siRNA directed to sequences common to rat, mouse and human Syk kinase RNA.
  • the expression of Syk kinase protein was analyzed by Western blotting using anti-Syk kinase Ab ( FIGS. 2 and 3 ).
  • Inhibition of Syk kinase expression in RBL cells treated with siRNA is shown in FIG. 2 , and levels of Syk kinase protein, normalized to levels of actin protein, are presented in Table 1.
  • Actin is a common protein used as a standard to examine protein expression.
  • Syk kinase protein expression in siRNA-treated RBL cells was inhibited by 45-51% ( FIG.
  • siRNA duplexes can comprise 21-nucleotide sense and 21-nucleotide antisense strands, paired in a manner to have a 2-nucleotide (dT) 3′ overhang.
  • the targeted region for siRNAs can be the sequence AA(N19) (N, any nucleotide) selected from the designated cDNA sequence beginning 50 to 100 nucleotides downstream of the start codon. G/C-contents of approximately 50% are preferred. Since expression of RNAs from pol III promoters is only efficient when the first transcribed nucleotide is a purine, it is preferred that the sense and antisense siRNAs begin with a purine nucleotide so that they can be expressed from pol III expression vectors without a change in targeting site.
  • siRNA targets in human Syk kinase mRNA were selected and then the sequences in rat and mouse Syk kinase mRNA were scanned in order to identify common Syk kinase mRNA target sequences in these species. Two appropriate target sequences were identified: Targeted region (1) (cDNA): 5′aagaagcccttcaaccggccc; Targeted region (2) (cDNA): 5′aacctcatcagggaatatctg.
  • RNA is not highly susceptible to nuclease degradation and the use of deoxynucleotides (thymidine (T) rather than uridine (U)) may affect the stability of the 3′ overhang.
  • T thymidine
  • U uridine
  • HS-24 cells (2 ⁇ 10 5 ) were pre-treated with 3 ⁇ l of siRNA-1 (control) DNA Target: 5′ AAGAAGCCCTTCAACCGGCCC Sense siRNA 5′- gaagcccuucaaccggccc dTdT 3′ Antisense 3′-dTdT cuucgggaaguuggccggg 5′ siRNA or siRNA-2 DNA Target: 5′ AACCTCATCAGGGAATATCTG sense 5′- ccucaucagggaauaucug dTdT 3′ antisense 3′-dTdT ggaguagucccuuauagac 5′ or Syk antisense, with Lipofectamine 2000 in a 12-well plate for 24 or 48 hr, and stimulated by 10 ng/ml of TNF overnight.
  • IL-6 in culture supernatant ELISA
  • ICAM-1 Flow cytometry
  • HS-24 cells were transiently transfected with siRNA-2 or siRNA-1 (control). Cell surface expression of ICAM-1, as well as release of IL-6, were then examined. Forty-eight-hour treatment of HS-24 cells with siRNA-2, but not with siRNA-1, significantly suppressed both Syk protein ( FIG. 5A ) and mRNA ( FIG. 5B ) expression.
  • HS-24 cells constitutively expressed low levels of ICAM-1 ( FIG. 6A ) not affected by siRNA-2 treatment (not shown).
  • Stimulation of HS-24 cells with 10 ng/ml of TNF during overnight culture caused significant increase in ICAM-1 expression both in resting cells (plated on poly-L-lysine) and cells adherent to fibronectin ( FIG. 6A ).
  • Cells adherent to fibronectin showed higher expression of ICAM-1 following stimulation with TNF compared to cells adherent to poly-L-lysine (P ⁇ 0.05).
  • Transfection with siRNA-2 down-regulated TNF-induced ICAM-1 expression in fibronectin-plated HS-24 cells (P ⁇ 0.005), but had no significant effect on ICAM-1 in poly-L-lysine-plated cells.
  • siRNA targeted to Syk kinase was studied in vivo in a Brown Norway rat model of ovalbumin (OA)-induced asthma.
  • Brown Norway rats were sensitized to OA i.p. as described (Laberge et al, Am. J. Respir. Crit. Care Med. 151:822 (1995)) and used on day 21 following sensitization.
  • siRNA was used alone without liposome or was used after formation of siRNA/liposome complexes.
  • 1,2 Dioleoyl-3-trimethylammonium-propane (DOTAP)/dioleoyl-phosphatidyl-ethanol-amine (DOPE) liposomes were prepared as previously described (see Stenton et al, J. Immunol. 169:1028 (2002) and references cited therein).
  • Cationic DOTAP:DOPE liposomes were incubated at a 2.5:1 ratio of the liposome to the siRNA and 125 micrograms of siRNA (with or without liposomes) was administered by aerosol following nebulization.
  • Bronchoalveolar lavage was carried out as described by Stenton et al, J. Immunol. 169:1028 (2002).
  • the isolated BAL cells were counted and cell smears were prepared by Cytospin. Cell differentials were counted in a blinded fashion following staining with HEMA-3 reagent (Biochemical Sciences, Swedesboro, N.J.).

Abstract

The present invention relates, in general, to Syk kinase and, in particular, to a method of inhibiting Syk kinase expression using small interfering RNA (siRNA).

Description

  • This application claims priority from Provisional Application No. 60/484,299, filed Jul. 3, 2003, the entire contents of which are incorporated herein by reference.
  • TECHNICAL FIELD
  • The present invention relates, in general, to Syk kinase and, in particular, to a method of inhibiting Syk kinase expression using small interfering RNA (siRNA).
  • BACKGROUND
  • Double stranded RNA has been shown to be a powerful agent for interfering with gene expression in a number of organisms, including C. elegans and Drosophila, as well as plants (Bernstein et al, RNA 7:1509-2151 (2001), McManus et al, Nat. Rev. Genet. 3:737-747 (2992), Hutvagner et al, Curr. Opin. Genet. Dev. 12:225-232 (2002), Zamore, Nat. Struct. Biol. 8:746-750 (2001) Tuschl et al, Genes Dev. 13:3191-3197 (1999)). Early problems in silencing mammalian genes with double stranded RNA arose because the mammalian immune system destroys cells that contain double stranded RNA, through mechanisms such as the interferon response, evolved as defense against invading RNA viruses (Clarke et al, RNA 1:7-20 (1995)). It has been demonstrated, however, that very short RNA fragments (e.g., 20-23 nt in length), designated small interfering RNA (siRNA), are able to escape the immune response. Thus introduced siRNAs can function well as gene silencing agents in mammalian cells (Elbashir et al, Nature 411:494-498 (2001), Elbashir et al, Genes Dev. 15:188-200 (2001), Paddison et al, Genes Dev. 16:948-958 (2002), Wianny et al, Nat. Cell Biol. 2:70-75 (2000)).
  • As it is presently understood, RNAi involves a multi-step process. Double stranded RNAs are cleaved by the endonuclease Dicer to generate 21-23 nucleotide fragments (siRNA). The siRNA duplex is resolved into 2 single stranded RNAs, one strand being incorporated into a protein-containing complex where it functions as guide RNA to direct cleavage of the target RNA (Schwarz et al, Mol. Cell. 10:537-548 (2002), Zamore et al, Cell 101:25-33 (2000)), thus silencing a specific genetic message (see also Zeng et al, Proc. Natl. Acad. Sci. 100:9779 (2003)).
  • Anti-sense DNA has also been widely used to inhibit gene expression (Roth et al, Annu. Rev. Biomed. Eng. 1:265-297 (1999)). Once inside the cell, anti-sense oligonucleotides (ASO) recognize, then bind tightly to complementary mRNA, thus preventing the mRNA from interacting with the protein translation machinery of the cell.
  • It has been demonstrated that inhibition of Syk kinase expression by Syk kinase mRNA ASO dramatically diminishes Fcγ receptor signaling (Matsuda et al, Molec. Biol. of the Cell 7:1095-1106 (1996)), and that Syk kinase mRNA ASO introduced by aerosol into rat lungs protects against Fcγ receptor-induced lung inflammation (Stenton et al, J. Immunol. 169:1028-1036 (2002)).
  • At least in certain systems, siRNA is more potent and reliable than ASO as an inhibitor of gene expression. The present invention results from studies designed to test the efficacy of siRNA as an inhibitor of Syk kinase expression.
  • SUMMARY OF THE INVENTION
  • The present invention relates generally to Syk kinase. In a preferred embodiment, the invention relates to a method of inhibiting Syk kinase expression using small interfering RNA (siRNA) and to therapeutic strategies based on such a method.
  • Objects and advantages of the present invention will be clear from the description that follows.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. The sense strand of each Syk kinase siRNA is the same sequence as the target sequence with the exception of the initial template adenine dimer and terminal overhang thymidine dimer. The antisense strand of the siRNA is the reverse complement of the target sequence.
  • FIG. 2. Expression of Syk kinase in RBL-2H3 cells transfected with siRNA targeted to Syk kinase mRNA. RBL-2H3 cells were transfected with siRNA-1 (Lane 2), siRNA-2 (lane 3) or lipofectamine transfection control (Lane 1). Proteins in cell lysates were separated by SDS-PAGE and transferred to nitrocellulose. Top panel, Syk kinase immunoblot; bottom panel, actin immunoblot.
  • FIG. 3. Expression of Syk kinase in human monocytes transfected with siRNA targeted to Syk kinase mRNA. Monocytes were treated with siRNA (Lane 2) or lipofectamine transfection control (Lane 1). Proteins in cell lysates were separated by SDS-PAGE, transferred to nitrocellulose and immunoblotted with anti-Syk kinase antibody.
  • FIG. 4. Western blot analysis: Syk protein expression in HS-24 cells.
  • FIGS. 5A and 5B. (FIG. 5A) HS-24 cells, following siRNA treatment, were lysed and equal amounts of total protein in HS-24 cell lysates were resolved by 10% SDS gel electrophoresis, and analyzed by Western blot using monoclonal antibody to Syk or actin. Lane 1—no treatment, lane 2—siRNA-1 (control) 24 h treatment, lane 3—siRNA-1 48 h treatment, lane 4—siRNA-2 24 h treatment, lane 5—siRNA-2 48 h treatment. (FIG. 5B) RNA was isolated and RT-PCR was performed for Syk and β-actin. Lane 1—no treatment, lane 2—siRNA-1 (control) 48 h treatment, lane 3—siRNA-2 48 h treatment.
  • FIGS. 6A and 6B. HS-24 cells plated on either polylysine coated plates (non stimulated, resting) or fibronectin coated plates (stimulated) were treated with siRNA-2, or siRNA-1 (control), or piceatannol. Cells were treated with 10 ng/ml of TNF during overnight culture. (FIG. 6A) Following siRNA (48 h) or piceatannol (16 h) treatment, cells were removed, immunostained with anti-CD54 (ICAM-1) and analysed by flow cytometry. (FIG. 6B) Cell culture supernatants were analysed for IL-6 release using an IL-6 ELISA kit. *P<0.05, **P<0.005 as compared to untreated cells (e.g., untreated with siRNA) stimulated with TNF. Results are representative of three to five independent experiments. The data indicate that inhibition of Syk expression by siRNA-2 down-regulates TNF-induced ICAM-1 expression and IL-6 release, important in the inflammatory response.
  • FIGS. 7A and 7B. Effect of siRNA targeted to Syk kinase delivered via aerosol on total cell numbers in bronchoalveolar lavage (BAL) fluid of ovalbumin (OA) sensitized and challenged Brown Norway Rats after three treatments. (FIG. 7A provides data as bar graphs, FIG. 7B shows individual data points (individual animals).
  • FIGS. 8A-8D. Effect of siRNA targeted to Syk kinase delivered via aerosol on numbers of macrophages, neutrophils, lymphocytes and eosinophils in BAL fluid of OA sensitized and challenged Brown Norway rats after three treatments. (FIG. 8A provides data as bar graphs, FIG. 8B shows individual data points (individual animals) for macrophage numbers, FIG. 8C shows individual data points for neutrophil numbers, and FIG. 8D shows individual data points for eosinophil numbers.)
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to RNA molecules that target Syk kinase mRNA. For example, the invention relates to RNA molecules about 19, 20 or 21 to about 23 nucleotides in length that direct cleavage and/or degradation of Syk kinase mRNA.
  • In a preferred embodiment, the present invention relates to the use of siRNA molecules, double stranded RNA molecules typically comprising two 20-23 nucleotide (nt) strands. SiRNAs suitable for use in the invention can be produced using any of a variety of approaches. The siRNA can be prepared in vitro and then introduced directly into cells (for example, by transfection). Alternatively, intracellular expression can be effected by transfecting into cells constructs (e.g., DNA-based vectors or cassettes) that express siRNA within cells.
  • More specifically, siRNA suitable for use in the invention can be prepared, for example, via chemical synthesis, in vitro transcription, enzymatic digestion of a longer dsRNA using an RNase III enzyme such as Dicer or RNase III, expression in cells from an siRNA expression plasmid or viral vector, or expression in cells from a PCR-derived siRNA expression cassette. Detailed descriptions of these various approaches are readily available and can be found, for example, at http://www.ambion.com/techlib/tn/103/2.html, www.bdbiosciences.com, www.oligoengine.com, www.genetherapysystems.com, www.dharmacon.com, http://www.mpibpc.gwdg.de/abteilungen/100/105/sirna.html, and/or in the references cited therein (which references are also incorporated herein by reference). (See also Sui et al, Proc Natl Acad Sci USA 99: 5515-20 (2002), Brummelkamp et al, Science 296:550-3 (2002),Paul et al, Nature Biotechnology 20:505-8 (2002), Lee et al, Nature Biotechnology 20: 500-5 (2002), Castanotto et al, RNA 8: 1454-60 (2002) and US Appln. 20030108923.)
  • Various approaches are available to enhance stability of RNA of the invention, (see, for example, U.S. Application Nos. 20020086356, 20020177570 and 20020055162, and U.S. Pat. Nos. 6,197,944, 6,590,093, 6,399,307, 6,057,134, 5,939,262, and 5,256,555, and references cited therein).
  • As indicated above, siRNA suitable for use in the invention can be prepared chemically. Advantageously, 2′ hydroxyls are protected during the synthetic process against degradation using, for example, acid labile orthoester protecting groups (see Scaringe et al, J. Am. Chem. Soc. 120:11820 (1998) and www.dharmacon.com (e.g., the ACE technology described therein)). The RNA oligomers can be simultaneously 2′ deprotected and annealed prior to use.
  • In chemically synthesized siRNA, at least one strand of the double stranded molecule can have a 3′ overhang from about 1 to about 6 nucleotides (e.g., pyrimidine and/or purine nucleotides) in length. Preferably, the 3′ overhang is from about 1 to about 5 nucleotides (e.g., thymidines or uridines), more preferably from about 1 to about 4 nucleotides and most preferably 2 or 3 nucleotides in length. Advantageously, each strand has an overhang. The length of the overhangs can be the same or different for each strand. Typically, both strands have overhangs of the same length. In a particular embodiment, the RNA of the present invention comprises 21 or 22 nucleotide strands that are paired and that have overhangs of from about 1 to about 3, particularly, about 2, nucleotides on the 3′ ends of both of the RNA strands.
  • As indicated above, siRNAs suitable for use in the invention can be prepared by enzymatic digestion of a longer dsRNA using an RNase III type enzyme (e.g., Dicer). (See references and web sites cited above.) For example, a commercially available Dicer siRNA generation kit can be used that permits generation of large numbers of siRNAs from full length target genes (Gene Therapy Systems, Inc, MV062603). SiRNA can be produced from target DNA and T7 RNA polymerase promoter sequences using PCR based cloning. Following RNA transcription from the target sequence, recombinant Dicer can cleave the transcribed RNAi into 22 bp siRNAs.
  • Also as indicated above, siRNA molecules suitable for use in the present invention can also be recombinantly produced using methods known in the art. (See references and web sites cited above.) Recombinant technology permits in vivo transcription of siRNAs in mammalian cells. In accordance with this approach, vectors can be used that contain, for example, RNA polymerase III or U6 promoter sequences. Such vectors (including viral vectors and plasmid vectors (such as pSIREN)) can be used as expression vectors or as shuttle vectors in conjunction with viral systems (e.g., adenoviral or retroviral systems) to introduce siRNA into mammalian cells. Vectors can be engineered to express sense and anti-sense strands of siRNAs that anneal in vivo to produce functional siRNAs. Alternatively, hairpin RNA can be expressed by inserting into a vector the sense strand (e.g., about 20 nt) of the target, followed by a short spacer (e.g., about 4 to about 10 nt), then the antisense strand of the target (e.g., about 20 nt) and, for example, about 5-6 T's as transcription terminator. The resulting RNA transcript folds back to form a stem-loop structure comprising, for example, about a 20 bp stem and about a 10 nt loop with 2-3 U's at the 3′ end. (See also Paddison et al (Proc. Natl. Acad. Sci. 99:1443-1448 (2002).) Constructs suitable for use in effecting in vivo production (including selection of vectors and promoters) can be readily designed by one skilled in the art and will vary, for example, with the cell/tissue target and the effect sought.
  • dsRNA can be used in the methods of the present invention provided it has sufficient homology to the targeted Syk kinase mRNA. SiRNA duplexes can be designed, for example, by searching Syk kinase cDNA for the target motif “AA(N)19”, wherein N is any nucleotide, motifs with approximately 30% to 70% G/C content being preferred, those of about 50% G/C content being more preferred. The sense strand of the siRNA duplex can correspond to nucleotides 3 to 21 of the selected AA(N)19 motif. The antisense strand of the siRNA duplex can have a sequence complementary to nucleotides 1 to 21 of the selected AA(N)19 motif. Further design details are provided at http://www.mpibpc.gwdg.de/abteilungen/100/105/sirna.html.
  • Preferred target sequences include sequences unique to Syk kinase mRNA. For example, target sequences can be selected from sequences between the two SH2 domains of Syk kinase or between the second SH2 domain and the kinase domain. Certain specific DNA target sequences are described in the non-limiting Examples that follow. Additional targets include, but are not limited to, the following:
    Identified homologies of
    *Sequence % GC 16-18/19 nucleotides
    AATATGTGAAGCAGACATGGA 42 mitochondrial ribosomal
    prot15
    AATCAAATCATACTCCTTCCC 42
    AAGAGAGTACTGTGTCATTCA 42
    AAGGAAAACCTCATCAGGGAA 47 inositolhexaphosphate
    kinase, β globin on
    Chr11
    AATCATACTCCTTCCCAAAGC 47
    AATTTTGGAGGCCGTCCACAA 53 oxytokinase
    AAGACTGGGCCCTTTGAGGAT 58
    AAGCACACATGGAACCTGCAG 58 histamine receptor H3,
    GTP binding protein
    AACTTCCAGGTTCCCATCCTG 58
    AAGCCTGGCCACAGAAAGTCC 63
    AAGCCCTACCCATGGACACAG 63
    AACCTGCAGGGTCAGGCTCTG 68
    AAGGGGTGCAGCCCAAGACTG 68 γ glutamyl transferase,
    rb prot L27a
    AACTTGCACCCTGGGCTGCAG 68 calcium channel α1E
    subunit
    AAGTCCTCCCCTGCCCAAGGG 74 NADH; ubiquinone oxido-
    reductase MLRQ subunit
    AAGGCCCCCAGAGAGAAGCCC 74
    AATCTCAAGAATCAAATCATA 26
    AATGTTAATTTTGGAGGCCGT 42
    AATCCGTATGAGCCAGAACTT 47
    AATCGGCACACAGGGAAATGT 53
    AACCGGCAAGAGAGTACTGTG 58
    AAGGAGGTTTACCTGGACCGA 58
  • The siRNAs described herein can be used in a variety of ways. For example, the siRNA molecules can be used to target Syk kinase mRNA in a cell or organism. In a specific embodiment, the siRNA can be introduced into human cells or a human in order to mediate RNAi in the cells or in cells in the individual, so as to prevent or treat a disease or undesirable condition associated with Syk kinase expression (e.g., inflammation of the lungs, joints eyes or bladder). The siRNA can also be used in the treatment of the immune destruction of blood cells, e.g., red blood cells in autoimmune hemolytic anemia and platelets in immune thrombocytopenic purpurea (ITP) (e.g., by targeting Syk kinase mRNA in macrophages and spleen and liver cells). In accordance with the instant method, the Syk kinase gene is targeted and the corresponding mRNA (the transcriptional product of the targeted Syk kinase gene) is degraded by RNAi. When lung cells are the target, an siRNA-containing composition can be aerosolized and administered, for example, via inhalation. Administration to joints can be effected by injection of an siRNA-containing solution. Administration to the eyes can be effected, for example, by injection or by application of drops comprising the siRNA in a carrier. Administration to the bladder, etc. can be effected, for example, by washing or irrigating the target tissue with a composition containing the siRNA. Administration to the skin can be via topical administration (e.g., as a liquid, cream or gel).
  • In accordance with the invention, cells of an individual (e.g., blood mononuclear cells, basophiles or mast cells) can be treated ex vivo so as to effect degradation of the Syk kinase mRNA. The cells to be treated can be obtained from the individual using known methods and the siRNAs that mediate degradation of the corresponding Syk kinase mRNA can be introduced into the cells, which can then be re-introduced into the individual.
  • In a specific embodiment, the invention relates to the use of the above-described siRNAs to inhibit mediator (e.g., histamine) release from cells bearing an Fcε receptor, such as mast cells. Inhibition of histamine (a mast cell mediator) release, for example, is of therapeutic importance in the treatment of asthma.
  • The siRNAs (or constructs suitable for use in effecting intracellular production of siRNA) of the invention can be administered systemically (e.g., via IV) or directly to the target tissue (e.g., via aerosol administration to the lung). Delivery can be effected using the techniques described herein (including liposome formulations). In addition to liposome formulations, polymer formulations can be used. Polyethylenimine (PEI) is an example of a suitable cationic polymer. Varying sizes of PEI can be used, including linear 22 kDa and branched 25 kDa PEI (other sizes, modified and unmodified, as well as biodegradable versions can be used). Delivery can also be effected using, for example, non-toxic viral delivery systems (e.g., an adeno-associated viral delivery system). Optimum dosing will depend on the patient, the siRNA, the mode of administration, and the effect sought. Optimum conditions can be established by one skilled in the art without undue experimentation.
  • Certain aspects of the invention can be described in greater detail in the non-limiting Examples that follows. (See also US Applications 20030084471, 20030108923 and 20020086356.)
  • EXAMPLE 1
  • Experimental Details
  • Reagents
  • Lipofectamine 2000 and Opti-Mem were purchased from Invitrogen (San Diego, Calif.). Eagle's MEM (EMEM), FCS, penicillin, and streptomycin were purchased from Life Technologies (Grand Island, N.Y.). Rabbit anti-murine Syk kinase polyclonal antibodies (Ab) and anti-actin Ab were purchased from Santa Cruz Biotechnology (Santa Cruz, Calif.), and F(ab′)2 goat anti-rabbit Ab was supplied by The Jackson Laboratory (Bar Harbor, Me.). Chemiluminescence reagent was purchased from DuPont NEN (Boston, Mass.).
  • Cells and Cell Lines
  • Peripheral blood monocytes obtained from healthy volunteers at the University of Pennsylvania were isolated as previously described (Matsuda et al, Molec. Biol. of the Cell 7:1095-1106 (1996)). Briefly, heparinized blood was centrifuged on Ficoll-Hypaque (Lymphocyte Separation Medium; Organon Teknika, Durham, N.C.) and interface cells were resuspended in complete medium containing RPMI 1640 (GIBCO BRL, Grand Island, N.Y.), 10% heat inactivated-fetal calf serum (FCS) (Intergen, Purchase, N.Y.) and 2 mM L-glutamine. Cells were allowed to adhere at 37° C. for 30 min to tissue culture flasks precoated with FCS. After 45-90 min, nonadherent cells were removed by extensive washing in Hanks' balanced salt solution. Cells were harvested by vigorous agitation. Monocytes were routinely>98% viable as judged by Trypan Blue exclusion. Isolated monocytes were maintained in RPMI 1640 supplemented with L-glutamine (2 mM) and 10% heat inactivated FCS at 37° C. in 5% CO2.
  • Rat basophilic cells (RBL-2H3) were grown in EMEM supplemented with 17% FBS, 100 U penicillin, 100 μg/ml streptomycin, and 4 mM glutamine at 37° C. in 5% CO2.
  • siRNA Duplex Construction
  • siRNAs for Syk kinase were prepared by Dharmacon Research Inc. (Lafayette, Colo.). In designing the siRNAs according to the guidelines provided by the manufacturer, potential siRNA targets (19 nucleotides immediately downstream of AA pairs) in human Syk kinase RNA were first identified. These sites were then scanned in the sequences of rat and mouse Syk kinase RNA in order to identify common Syk kinase RNA target sequences in these species. Two appropriate sites were identified, and two 21-mer RNAs, each consisting of 19 complementary nucleotides and 3′ terminal noncomplementary dimers of thymidine (Elbashir et al, Nature 411:494-498 (2001)), were constructed. The sense strand of the siRNA is the same sequence as the target mRNA sequence with the exception of the terminal thymidine dimer. The antisense strand of the siRNA is the reverse complement of the target sequence.
    1) siRNA-1: Human, bp 296 to bp 316; Mouse and
    Rat, bp 307 to bp 327.
    sense 5′-     gaagcccuucaaccggccc dTdT 3′
    antisense 3′-dTdT cuucgggaaguuggccggg 5′
    2) siRNA-2: Human, bp 364 to bp 382; Mouse and
    Rat, bp 375 to bp 393
    sense 5′-     ccucaucagggaauaucug dTdT 3′
    antisense 3′-dTdT ggaguagucccuuauagac 5′

    Transfections
  • siRNA was introduced into RBL-2H3 cells and into monocytes by transfection. For the transfections, 5×104 RBL-2H3 cells or 1×105 monocytes were seeded into each well of a 24-well plate. Twenty-four hours later, the complete medium was replaced with 400 μl fresh medium lacking serum and antibiotic and siRNA/Lipofectamine 2000 complex was added to each well. For the RBL cells, the siRNA/Lipofectamine 2000 complex was formed by adding 3 μl of siRNA duplex (20 μM) and 3 μl of Lipofectamine 2000 to 100 μl Opti-mem without serum or antibiotic according to the manufacturer's protocol. For monocytes, the siRNA/Lipofectamine 2000 complex was formed by adding 3 μl of siRNA duplex (20 μM) and 1 μl of Lipofectamine 2000 to 100 μl Opti-mem without serum or antibiotic. Cells were incubated at 37° C. for 48 hours before examination of kinase protein expression by Western blotting.
  • Western Blot Analysis of Syk Kinase Protein
  • Lysates were prepared by boiling cells in Laemmli sample buffer (2% SDS, 10% glycerol, 100 mM DTT, and 60 mM Tris (pH 6.8) for 5 minutes. Proteins in lysates were separated by SDS-PAGE (10% polyacrylamide) and transferred to a nitrocellulose membrane in sample buffer (25 mM Tris, 190 mM glycine, and 20% methanol). The nitrocellulose membrane was incubated overnight at 4° C. with 1 μg/ml rabbit anti-murine Syk kinsae polyclonal Ab before incubation with goat anti-rabbit HRP (1.5 h at room temperature). Protein bands on the membrane were visualized with chemiluminescence reagent according to the manufacturer's protocol. After detection of Syk kinase protein, the anti-Syk kinase Ab was removed by incubating the membrane in a stripping buffer containing 100 mM 2-ME, 2% SDS, and 62.5 mM Tris-HCL (pH 6.7) for 30 minutes at 50° C. with occasional agitation. The membrane was then reprobed with anti-actin Ab and bands on the membrane were visualized with chemiluminescence reagent. Protein levels of Syk kinase were quantified by densitometry (Personal Densitometer, Molecular Dynamics).
  • Results
  • Effects of siRNA on the Expression of Syk Kinase
  • Rat basophilic cells (RBL-2H3 cells) and human monocytes were transfected with siRNA directed to sequences common to rat, mouse and human Syk kinase RNA. The expression of Syk kinase protein was analyzed by Western blotting using anti-Syk kinase Ab (FIGS. 2 and 3). Inhibition of Syk kinase expression in RBL cells treated with siRNA is shown in FIG. 2, and levels of Syk kinase protein, normalized to levels of actin protein, are presented in Table 1. Actin is a common protein used as a standard to examine protein expression. Syk kinase protein expression in siRNA-treated RBL cells was inhibited by 45-51% (FIG. 2, lanes 2 and 3) compared to untreated RBL cells (lane 1). The magnitude of the inhibition of Syk kinase gene expression by siRNA in cultured cells was encouraging since previous experiments with Syk kinase mRNA ASO indicated that greater levels of Syk kinase inhibition can be achieved in non-multiplying cells such as monocytes than in multiplying cells cultured in vitro. Inhibition of Syk kinase expression by siRNA in monocytes maintained in culture (FIG. 3) was also observed. Syk kinase expression in siRNA-treated monocytes was inhibited (FIG. 3, lane 2). In addition, Syk kinase expression in siRNA-treated U937 cells and siRNA-treated THP-1 cells was inhibited (U937 and THP-1 being macrophage-like cell lines). The data thus demonstrate the effectiveness of siRNA directed against Syk kinase RNA to suppress the expression of this gene and indicate that siRNA directed against Syk kinase RNA can serve as a powerful therapeutic tool to combat inflammation.
    TABLE 1
    Densitometry quantification of Syk
    kinase expression in RBL cells.
    Transfection Syk kinase Syk kinase
    Control siRNA1 siRNA2
    Syk* 2040 953 627
    Syk Corr** 1114 997
    % Syk Inhibition 0 45 51

    *densitometry units

    **Syk densitometry units corrected for % actin expression in each lane.

    Discussion
  • The siRNAs used in these studies were chemically synthesized (Dharmicon Research Inc., Lafayette, Colo.) but siRNAs can also be prepared by recombinant techniques. siRNA duplexes can comprise 21-nucleotide sense and 21-nucleotide antisense strands, paired in a manner to have a 2-nucleotide (dT) 3′ overhang.
  • The targeted region for siRNAs can be the sequence AA(N19) (N, any nucleotide) selected from the designated cDNA sequence beginning 50 to 100 nucleotides downstream of the start codon. G/C-contents of approximately 50% are preferred. Since expression of RNAs from pol III promoters is only efficient when the first transcribed nucleotide is a purine, it is preferred that the sense and antisense siRNAs begin with a purine nucleotide so that they can be expressed from pol III expression vectors without a change in targeting site.
  • In the studies described above, potential siRNA targets in human Syk kinase mRNA were selected and then the sequences in rat and mouse Syk kinase mRNA were scanned in order to identify common Syk kinase mRNA target sequences in these species. Two appropriate target sequences were identified:
    Targeted region (1) (cDNA):
    5′aagaagcccttcaaccggccc;
    Targeted region (2) (cDNA):
    5′aacctcatcagggaatatctg.
  • The target sequences and Syk kinase siRNAs are shown in FIG. 1. Duplexed RNA is not highly susceptible to nuclease degradation and the use of deoxynucleotides (thymidine (T) rather than uridine (U)) may affect the stability of the 3′ overhang.
  • EXAMPLE 2
  • HS-24 cells (2 × 105) were pre-treated with 3 μl
    of siRNA-1 (control)
    DNA Target: 5′ AAGAAGCCCTTCAACCGGCCC
    Sense siRNA
    5′-     gaagcccuucaaccggccc dTdT 3′
    Antisense 3′-dTdT cuucgggaaguuggccggg 5′
    siRNA
    or
    siRNA-2
    DNA Target: 5′ AACCTCATCAGGGAATATCTG
    sense
    5′-     ccucaucagggaauaucug dTdT 3′
    antisense 3′-dTdT ggaguagucccuuauagac 5′

    or Syk antisense, with Lipofectamine 2000 in a 12-well plate for 24 or 48 hr, and stimulated by 10 ng/ml of TNF overnight. Read-outs: IL-6 in culture supernatant (ELISA) and cell surface expression of ICAM-1 (Flow cytometry). As shown by Western blot analysis (see FIG. 4), siRNA-2 caused a decrease in Syk protein expression following 48 hr of treatment.
  • More specifically, HS-24 cells were transiently transfected with siRNA-2 or siRNA-1 (control). Cell surface expression of ICAM-1, as well as release of IL-6, were then examined. Forty-eight-hour treatment of HS-24 cells with siRNA-2, but not with siRNA-1, significantly suppressed both Syk protein (FIG. 5A) and mRNA (FIG. 5B) expression.
  • HS-24 cells constitutively expressed low levels of ICAM-1 (FIG. 6A) not affected by siRNA-2 treatment (not shown). Stimulation of HS-24 cells with 10 ng/ml of TNF during overnight culture caused significant increase in ICAM-1 expression both in resting cells (plated on poly-L-lysine) and cells adherent to fibronectin (FIG. 6A). Cells adherent to fibronectin showed higher expression of ICAM-1 following stimulation with TNF compared to cells adherent to poly-L-lysine (P<0.05). Transfection with siRNA-2 down-regulated TNF-induced ICAM-1 expression in fibronectin-plated HS-24 cells (P<0.005), but had no significant effect on ICAM-1 in poly-L-lysine-plated cells. Overnight treatment with the pharmacological Syk inhibitor, piceatannol (10 μM), caused significant down-regulation of ICAM-1 in TNF-stimulated cells both in poly-L-lysine and fibronectin adherent conditions (FIG. 6A). As determined by trypan blue dye exclusion, the treatment of cells with siRNA or piceatannol had no significant effect on viability (in all experiments, viability was >96%).
  • Although IL-6 release by HS-24 cells without TNF stimulation was minimal, there was a trend to higher IL-6 levels in culture supernatants of cells adherent to fibronectin (FIG. 6B). As expected, a great elevation of IL-6 levels was observed following TNF stimulation in both culture conditions, with higher levels in fibronectin-adherent cells (P<0.05). siRNA-2 treatment caused down-regulation of IL-6 release (55-58%) that reached statistical significance in fibronectin-adherent culture (P<0.05). Piceatannol almost completely inhibited TNF-induced IL-6 release (FIG. 6B).
  • Thus, inhibition of Syk kinase down-regulated TNF-induced expression of ICAM-1 and IL-6 release, hallmarks of inflammatory responses in the airway epithelium. The effect was significant in cells adherent to fibronectin indicating that Syk involvement in these pro-inflammatory events is at least partly β1 integrin dependent.
  • EXAMPLE 3
  • The effects of siRNA targeted to Syk kinase was studied in vivo in a Brown Norway rat model of ovalbumin (OA)-induced asthma.
  • Brown Norway rats were sensitized to OA i.p. as described (Laberge et al, Am. J. Respir. Crit. Care Med. 151:822 (1995)) and used on day 21 following sensitization.
  • The siRNA used in this Example is as follows:
    DNA Target: 5′ AACCTCATCAGGGAATATCTG
    sense
    5′-   ccucaucagggaauaucug uu 3′
    antisense 3′-uu ggaguagucccuuauagac    5′

    The description siRNA 2M, used in FIGS. 7 and 8, refers to the above, modified by Dharmacon to provide additional stability. The designation siRNA-2, used in FIGS. 7 and 8, refers to the above sequence in unmodified form.
  • siRNA was used alone without liposome or was used after formation of siRNA/liposome complexes. 1,2 Dioleoyl-3-trimethylammonium-propane (DOTAP)/dioleoyl-phosphatidyl-ethanol-amine (DOPE) liposomes were prepared as previously described (see Stenton et al, J. Immunol. 169:1028 (2002) and references cited therein). Cationic DOTAP:DOPE liposomes were incubated at a 2.5:1 ratio of the liposome to the siRNA and 125 micrograms of siRNA (with or without liposomes) was administered by aerosol following nebulization.
  • The aerosolized administration of Syk kinase-targeted siRNA was as described by Stenton et al, J. Immunol. 164:3790 (2000). Nine milliters of saline, siRNA or siRNA/liposome were administered per rat by nebulization for 45 min using a Sidestream nebulizer as described in Stenton et al, J. Immunol. 169:1028 (2002). Twenty-four hours later, the procedure was repeated, followed by a third treatment at 48 h. Immediately after the third treatment, rats were challenged with aerosolized saline or 5% OA in saline for 5 min. Twenty-four hours after challenge, the animals were sacrificed.
  • Bronchoalveolar lavage (BAL) was carried out as described by Stenton et al, J. Immunol. 169:1028 (2002).
  • The isolated BAL cells were counted and cell smears were prepared by Cytospin. Cell differentials were counted in a blinded fashion following staining with HEMA-3 reagent (Biochemical Sciences, Swedesboro, N.J.).
  • Summarizing, the Brown Norway rat model of OA induced allergic asthma and pulmonary inflammation was used in the above-described studies. Pulmonary inflammation, as determined by recruitment of cells to BAL fluid, was dramatically inhibited by Syk kinase-targeted siRNA in the presence and absence of liposomes.
  • All documents and other information sources cited above are hereby incorporated in their entirety by reference.

Claims (5)

1. A method of inhibiting expression of Syk kinase in a cell comprising introducing into said cell small interfering RNA (siRNA) molecules that direct cleavage of a target Syk kinase mRNA sequence present in said cell thereby effecting said inhibition.
2-18. (canceled)
19. An siRNA molecule comprising a sequence complementary to a sequence selected from the group consisting of:
AATATGTGAAGCAGACATGGA, AATCAAATCATACTCCTTCCC, AAGAGAGTACTGTGTCATTCA, AAGGAAAACCTCATCAGGGAA, AATCATACTCCTTCCCAAAGC, AATTTTGGAGGCCGTCCACAA, AAGACTGGGCCCTTTGAGGAT, AAGCAGACATGGAACCTGCAG, AACTTCCAGGTTCCCATCCTG, AAGCCTGGCCACAGAAAGTCC, AAGCCCTACCCATGGACACAG, AACCTGCAGGGTCAGGCTCTG, AAGGGGTGCAGCCCAAGACTG, AACTTGCACCCTGGGCTGCAG, AAGTCCTCCCCTGCCCAAGGG, AAGGCCCCCAGAGAGAAGCCC, AATCTCAAGAATCAAATCATA, AATGTTAATTTTGGAGGCCGT, AATCCGTATGAGCCAGAACTT, AATCGGCACACAGGGAAATGT, AACCGGCAAGAGAGTACTGTG, AAGGAGGTTTACCTGGACCGA, and AACCTCATCAGGGAATATCTG.
20. A composition comprising the siRNA molecule according to claim 19 and a carrier.
21. A composition comprising the siRNA molecule according to claim 19 and a liposome or polymer.
US11/639,243 2003-07-03 2006-12-15 Inhibition of Syk kinase expression Abandoned US20070219152A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/639,243 US20070219152A1 (en) 2003-07-03 2006-12-15 Inhibition of Syk kinase expression
US13/067,035 US20120093913A1 (en) 2003-07-03 2011-05-03 Inhibition of SYK kinase expression

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US48429903P 2003-07-03 2003-07-03
US10/880,612 US7173015B2 (en) 2003-07-03 2004-07-01 Inhibition of Syk kinase expression
US11/639,243 US20070219152A1 (en) 2003-07-03 2006-12-15 Inhibition of Syk kinase expression

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/880,612 Division US7173015B2 (en) 2003-07-03 2004-07-01 Inhibition of Syk kinase expression

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US65678510A Continuation 2003-07-03 2010-02-16

Publications (1)

Publication Number Publication Date
US20070219152A1 true US20070219152A1 (en) 2007-09-20

Family

ID=34079054

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/880,612 Expired - Fee Related US7173015B2 (en) 2003-07-03 2004-07-01 Inhibition of Syk kinase expression
US11/639,243 Abandoned US20070219152A1 (en) 2003-07-03 2006-12-15 Inhibition of Syk kinase expression
US13/067,035 Abandoned US20120093913A1 (en) 2003-07-03 2011-05-03 Inhibition of SYK kinase expression

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/880,612 Expired - Fee Related US7173015B2 (en) 2003-07-03 2004-07-01 Inhibition of Syk kinase expression

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/067,035 Abandoned US20120093913A1 (en) 2003-07-03 2011-05-03 Inhibition of SYK kinase expression

Country Status (8)

Country Link
US (3) US7173015B2 (en)
EP (2) EP1692153A4 (en)
JP (2) JP2007524397A (en)
CN (2) CN100577680C (en)
AU (1) AU2004257167B2 (en)
CA (1) CA2531069A1 (en)
HK (1) HK1096972A1 (en)
WO (1) WO2005007623A2 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009061368A1 (en) * 2007-11-06 2009-05-14 Benaroya Research Institute Inhibition of versican with sirna and other molecules
US20110124567A1 (en) * 2000-04-13 2011-05-26 Wight Thomas N Therapeutic compounds and methods
WO2011100341A1 (en) * 2010-02-09 2011-08-18 Drexel University New signaling molecule involved in ultraviolet damage to skin
WO2014071109A1 (en) 2012-11-01 2014-05-08 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
WO2014151386A1 (en) 2013-03-15 2014-09-25 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
WO2014194254A1 (en) 2013-05-30 2014-12-04 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
WO2015051244A1 (en) 2013-10-04 2015-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015051241A1 (en) 2013-10-04 2015-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015061204A1 (en) 2013-10-21 2015-04-30 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015143012A1 (en) 2014-03-19 2015-09-24 Infinity Pharmaceuticals, Inc. Heterocyclic compounds for use in the treatment of pi3k-gamma mediated disorders
WO2015160986A2 (en) 2014-04-16 2015-10-22 Infinity Pharmaceuticals, Inc. Combination therapies
WO2016054491A1 (en) 2014-10-03 2016-04-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2017214269A1 (en) 2016-06-08 2017-12-14 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2017223422A1 (en) 2016-06-24 2017-12-28 Infinity Pharmaceuticals, Inc. Combination therapies

Families Citing this family (112)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060160759A1 (en) * 2002-09-28 2006-07-20 Jianzhu Chen Influenza therapeutic
US7368559B2 (en) 2003-11-14 2008-05-06 Diana Beardsley FcγRIIA-specific nucleic acid interference
CA2576925C (en) 2004-08-16 2013-12-10 The Cbr Institute For Biomedical Research, Inc. Method of delivering rna interference and uses thereof
US7507809B2 (en) * 2005-01-07 2009-03-24 Alnylam Pharmaceuticals, Inc. RNAi modulation of RSV and therapeutic uses thereof
WO2006113743A2 (en) * 2005-04-18 2006-10-26 Massachusetts Institute Of Technology Compositions and methods for rna interference with sialidase expression and uses thereof
TW201336514A (en) * 2006-04-13 2013-09-16 Alcon Res Ltd RNAi-mediated inhibition of spleen tyrosine kinase-related inflammatory conditions
US20080139531A1 (en) * 2006-12-04 2008-06-12 Alcon Manufacturing Ltd. Use of connective tissue mast cell stabilizers to facilitate ocular surface re-epithelization and wound repair
CN101641094A (en) * 2007-04-11 2010-02-03 爱尔康研究有限公司 Use of an inhibitor of tnfa plus an antihistamine to treat allergic rhinitis and allergic conjunctivitis
WO2008132723A2 (en) * 2007-04-26 2008-11-06 Quark Pharmaceuticals, Inc. Therapeutic delivery of inhibitory nucleic acid molecules to the respiratory system
WO2009033027A2 (en) * 2007-09-05 2009-03-12 Medtronic, Inc. Suppression of scn9a gene expression and/or function for the treatment of pain
EP2229459B1 (en) * 2007-12-13 2014-08-27 Alnylam Pharmaceuticals, Inc. Methods and compositions for prevention or treatment of RSV infection
EP2346883B1 (en) 2008-09-23 2016-03-23 Scott G. Petersen Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating rna interference
EP2350277A1 (en) * 2008-10-23 2011-08-03 Alnylam Pharmaceuticals, Inc. Methods and compositions for prevention or treatment of rsv infection using modified duplex rna molecules
US20110082145A1 (en) * 2009-10-01 2011-04-07 Alcon Research, Ltd. Olopatadine compositions and uses thereof
KR101223483B1 (en) * 2010-09-10 2013-01-17 한국과학기술연구원 NEW POLYMERIC NANO-PARTICLES FOR siRNA DELIVERY USING CHARGE INTERACTION AND COVALENT BONDING
AU2011336467A1 (en) 2010-12-01 2013-07-04 Spinal Modulation, Inc. Agent delivery systems for selective neuromodulation
TW201536329A (en) 2013-08-09 2015-10-01 Isis Pharmaceuticals Inc Compounds and methods for modulation of dystrophia myotonica-protein kinase (DMPK) expression
ES2921874T3 (en) 2014-02-28 2022-09-01 Nimbus Lakshmi Inc TYK2 inhibitors and uses thereof
EP3134733B1 (en) * 2014-04-25 2020-10-14 The Brigham and Women's Hospital, Inc. Assay and method for treating subjects with immune-mediated diseases
JP6749890B2 (en) 2014-08-12 2020-09-02 モナッシュ ユニバーシティ Lymphoid prodrug
TWI744225B (en) 2015-02-27 2021-11-01 美商林伯士拉克許米公司 Tyk2 inhibitors and uses thereof
WO2017004134A1 (en) 2015-06-29 2017-01-05 Nimbus Iris, Inc. Irak inhibitors and uses thereof
WO2017040757A1 (en) 2015-09-02 2017-03-09 Nimbus Lakshmi, Inc. Tyk2 inhibitors and uses thereof
EP3347340A4 (en) 2015-09-08 2019-01-23 Monash University Lymph directing prodrugs
US10683308B2 (en) 2015-09-11 2020-06-16 Navitor Pharmaceuticals, Inc. Rapamycin analogs and uses thereof
LT3364958T (en) 2015-10-23 2023-06-12 Navitor Pharmaceuticals, Inc. Modulators of sestrin-gator2 interaction and uses thereof
EP3389664A4 (en) 2015-12-14 2020-01-08 Raze Therapeutics Inc. Caffeine inhibitors of mthfd2 and uses thereof
HUE055197T2 (en) 2016-03-09 2021-11-29 Raze Therapeutics Inc 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
US11014882B2 (en) * 2016-03-09 2021-05-25 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
CA3019394A1 (en) 2016-04-08 2017-10-12 X4 Pharmaceuticals, Inc. Methods for treating cancer
ES2870920T3 (en) 2016-06-21 2021-10-28 X4 Pharmaceuticals Inc CXCR4 inhibitors and their uses
JP6994767B2 (en) 2016-06-21 2022-01-14 エックス4 ファーマシューティカルズ, インコーポレイテッド CXCR4 inhibitor and its use
CA3027495A1 (en) 2016-06-21 2017-12-28 X4 Pharmaceuticals, Inc. Cxcr4 inhibitors and uses thereof
US10323036B2 (en) 2016-10-14 2019-06-18 Nimbus Lakshmi, Inc. TYK2 inhibitors and uses thereof
AU2017345736B2 (en) 2016-10-21 2022-04-07 Takeda Pharmaceutical Company Limited TYK2 inhibitors and uses thereof
US10414727B2 (en) 2016-11-08 2019-09-17 Navitor Pharmaceuticals, Inc. Phenyl amino piperidine mTORC inhibitors and uses thereof
US20180193270A1 (en) 2016-11-29 2018-07-12 PureTech Health LLC Exosomes for delivery of therapeutic agents
US11091451B2 (en) 2016-12-05 2021-08-17 Raze Therapeutics, Inc. SHMT inhibitors and uses thereof
EP3559018A1 (en) 2016-12-23 2019-10-30 Bicyclerd Limited Peptide derivatives having novel linkage structures
WO2018127699A1 (en) 2017-01-06 2018-07-12 Bicyclerd Limited Compounds for treating cancer
EP3592746B1 (en) 2017-03-08 2024-01-24 Takeda Pharmaceutical Company Limited Tyk2 inhibitors, uses, and methods for production thereof
EP3375778A1 (en) 2017-03-14 2018-09-19 Artax Biopharma Inc. Aryl-piperidine derivatives
EP3375784A1 (en) 2017-03-14 2018-09-19 Artax Biopharma Inc. Aza-dihydro-acridone derivatives
WO2018191146A1 (en) 2017-04-10 2018-10-18 Navitor Pharmaceuticals, Inc. Heteroaryl rheb inhibitors and uses thereof
EP3615019A4 (en) 2017-04-26 2021-01-27 Navitor Pharmaceuticals, Inc. Modulators of sestrin-gator2 interaction and uses thereof
EP3615550A1 (en) 2017-04-27 2020-03-04 BicycleTx Limited Bicyclic peptide ligands and uses thereof
CN111032678A (en) 2017-06-26 2020-04-17 拜西克尔德有限公司 Bicyclic peptide ligands with detectable moieties and uses thereof
MX2020001103A (en) 2017-07-28 2020-08-17 Nimbus Lakshmi Inc Tyk2 inhibitors and uses thereof.
US20200291096A1 (en) 2017-08-14 2020-09-17 Bicyclerd Limited Bicyclic peptide ligand sting conjugates and uses thereof
US20200283482A1 (en) 2017-08-14 2020-09-10 Bicyclerd Limited Bicyclic peptide ligand prr-a conjugates and uses thereof
US11883497B2 (en) 2017-08-29 2024-01-30 Puretech Lyt, Inc. Lymphatic system-directing lipid prodrugs
AU2018324037A1 (en) 2017-08-29 2020-04-16 Monash University Lymphatic system-directing lipid prodrugs
US11358948B2 (en) 2017-09-22 2022-06-14 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
IL295603B2 (en) 2017-09-22 2024-03-01 Kymera Therapeutics Inc Protein degraders and uses thereof
TWI825046B (en) 2017-12-19 2023-12-11 英商拜西可泰克斯有限公司 Bicyclic peptide ligands specific for epha2
GB201721265D0 (en) 2017-12-19 2018-01-31 Bicyclerd Ltd Bicyclic peptide ligands specific for EphA2
US11608345B1 (en) 2017-12-19 2023-03-21 Puretech Lyt, Inc. Lipid prodrugs of rapamycin and its analogs and uses thereof
US11304954B2 (en) 2017-12-19 2022-04-19 Puretech Lyt, Inc. Lipid prodrugs of mycophenolic acid and uses thereof
WO2019126378A1 (en) 2017-12-19 2019-06-27 Ariya Therapeutics, Inc. Lipid prodrugs of mycophenolic acid and uses thereof
CN112105385A (en) 2017-12-26 2020-12-18 凯麦拉医疗公司 IRAK degrading agents and uses thereof
WO2019140380A1 (en) 2018-01-12 2019-07-18 Kymera Therapeutics, Inc. Protein degraders and uses thereof
EP3737675A4 (en) 2018-01-12 2022-01-05 Kymera Therapeutics, Inc. Crbn ligands and uses thereof
CN111918651B (en) 2018-01-29 2024-01-30 默克专利股份有限公司 GCN2 inhibitors and uses thereof
SG11202006832YA (en) 2018-01-29 2020-08-28 Merck Patent Gmbh Gcn2 inhibitors and uses thereof
IL301120A (en) 2018-02-27 2023-05-01 Artax Biopharma Inc Chromene derivatives as inhibitors of tcr-nck interaction
IL278142B2 (en) 2018-04-24 2023-11-01 Merck Patent Gmbh Antiproliferation compounds and uses thereof
EP3784669B1 (en) 2018-04-24 2023-10-25 Vertex Pharmaceuticals Incorporated Pteridinone compounds and uses thereof
CA3103282A1 (en) 2018-06-15 2019-12-19 Navitor Pharmaceuticals, Inc. Rapamycin analogs and uses thereof
US11180531B2 (en) 2018-06-22 2021-11-23 Bicycletx Limited Bicyclic peptide ligands specific for Nectin-4
GB201810316D0 (en) 2018-06-22 2018-08-08 Bicyclerd Ltd Peptide ligands for binding to EphA2
EP3817748A4 (en) 2018-07-06 2022-08-24 Kymera Therapeutics, Inc. Tricyclic crbn ligands and uses thereof
US10548889B1 (en) 2018-08-31 2020-02-04 X4 Pharmaceuticals, Inc. Compositions of CXCR4 inhibitors and methods of preparation and use
JP2022500499A (en) 2018-09-07 2022-01-04 ピク セラピューティクス, インコーポレイテッド EIF4E Inhibitors and Their Use
CN113271940A (en) 2018-10-15 2021-08-17 林伯士拉克许米公司 TYK2 inhibitors and uses thereof
EP3870597A1 (en) 2018-10-23 2021-09-01 BicycleTx Limited Bicyclic peptide ligands and uses thereof
US11345654B2 (en) 2018-10-24 2022-05-31 Navitor Pharmaceuticals, Inc. Polymorphic compounds and uses thereof
US11053241B2 (en) 2018-11-30 2021-07-06 Nimbus Lakshmi, Inc. TYK2 inhibitors and uses thereof
KR20210111252A (en) 2018-11-30 2021-09-10 카이메라 쎄라퓨틱스 인코포레이티드 IRAK disintegrants and uses thereof
EP3670659A1 (en) 2018-12-20 2020-06-24 Abivax Biomarkers, and uses in treatment of viral infections, inflammations, or cancer
EP3914357A4 (en) 2019-01-23 2022-10-12 Nimbus Lakshmi, Inc. Tyk2 inhibitors and uses thereof
WO2020165600A1 (en) 2019-02-14 2020-08-20 Bicycletx Limited Bicyclic peptide ligand sting conjugates and uses thereof
CA3135569A1 (en) 2019-04-02 2020-10-08 Bicycletx Limited Bicycle toxin conjugates and uses thereof
WO2020206424A1 (en) 2019-04-05 2020-10-08 Kymera Therapeutics, Inc. Stat degraders and uses thereof
WO2020243423A1 (en) 2019-05-31 2020-12-03 Ikena Oncology, Inc. Tead inhibitors and uses thereof
AU2020345962A1 (en) 2019-09-11 2022-03-31 Vincere Biosciences, Inc. USP30 inhibitors and uses thereof
EP4027995A4 (en) 2019-09-13 2023-08-23 Nimbus Saturn, Inc. Hpk1 antagonists and uses thereof
IL292612A (en) 2019-11-01 2022-07-01 Navitor Pharm Inc Methods of treatment using an mtorc1 modulator
WO2021113665A1 (en) 2019-12-05 2021-06-10 Navitor Pharmaceuticals, Inc. Rapamycin analogs and uses thereof
EP4076524A4 (en) 2019-12-17 2023-11-29 Kymera Therapeutics, Inc. Irak degraders and uses thereof
US11779578B2 (en) 2019-12-17 2023-10-10 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
AU2020412780A1 (en) 2019-12-23 2022-07-21 Kymera Therapeutics, Inc. SMARCA degraders and uses thereof
CN115515685A (en) 2020-03-03 2022-12-23 皮克医疗公司 EIF4E inhibitor and application thereof
TW202210483A (en) 2020-06-03 2022-03-16 美商凱麥拉醫療公司 Crystalline forms of irak degraders
CN116234931A (en) 2020-08-17 2023-06-06 拜斯科技术开发有限公司 Bicyclic conjugates with specificity for NECTIN-4 and uses thereof
AU2021392040A1 (en) 2020-12-02 2023-06-29 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2022120353A1 (en) 2020-12-02 2022-06-09 Ikena Oncology, Inc. Tead inhibitors and uses thereof
AU2022215844A1 (en) 2021-02-02 2023-09-14 Liminal Biosciences Limited Gpr84 antagonists and uses thereof
CA3206501A1 (en) 2021-02-02 2022-08-11 Shaun Abbott Gpr84 antagonists and uses thereof
CN116867494A (en) 2021-02-15 2023-10-10 凯麦拉医疗公司 IRAK4 degrading agent and use thereof
CN117295737A (en) 2021-03-05 2023-12-26 林伯士萨顿公司 HPK1 antagonists and uses thereof
KR20230172548A (en) 2021-04-16 2023-12-22 이케나 온콜로지, 인코포레이티드 MEK inhibitors and uses thereof
AU2022334296A1 (en) 2021-08-25 2024-03-07 PIC Therapeutics, Inc. Eif4e inhibitors and uses thereof
WO2023028238A1 (en) 2021-08-25 2023-03-02 PIC Therapeutics, Inc. Eif4e inhibitors and uses thereof
WO2023034870A2 (en) 2021-09-01 2023-03-09 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing dmpk expression
WO2023114984A1 (en) 2021-12-17 2023-06-22 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2023173057A1 (en) 2022-03-10 2023-09-14 Ikena Oncology, Inc. Mek inhibitors and uses thereof
WO2023173053A1 (en) 2022-03-10 2023-09-14 Ikena Oncology, Inc. Mek inhibitors and uses thereof
WO2023211889A1 (en) 2022-04-25 2023-11-02 Ikena Oncology, Inc. Polymorphic compounds and uses thereof
WO2023230205A1 (en) 2022-05-25 2023-11-30 Ikena Oncology, Inc. Mek inhibitors and uses thereof
WO2024028363A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Heteroaryl carboxamide and related gpr84 antagonists and uses thereof
WO2024028364A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Aryl-triazolyl and related gpr84 antagonists and uses thereof
WO2024028365A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Substituted pyridone gpr84 antagonists and uses thereof

Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6242427B1 (en) * 1993-09-30 2001-06-05 University Of Pennsylvania Methods of inhibiting phagocytosis
US20030232364A1 (en) * 2001-11-07 2003-12-18 Shaughnessy John D. Diagnosis, prognosis and identification of potential therapeutic targets of multiple myeloma based on gene expression profiling
US20040009523A1 (en) * 2001-11-07 2004-01-15 Shaughnessy John D. Diagnosis, prognosis and identification of potential therapeutic targets of multiple myeloma based on gene expression profiling
US20040018522A1 (en) * 2002-05-09 2004-01-29 Brigham And Women's Hospital, Inc. Identification of dysregulated genes in patients with multiple sclerosis
US20040028685A1 (en) * 2002-05-10 2004-02-12 Kinch Michael S. EphA2 monoclonal antibodies and methods of use thereof
US20040106156A1 (en) * 2002-07-10 2004-06-03 Perez Omar D. Methods and compositions for detecting receptor-ligand interactions in single cells
US20040219575A1 (en) * 2002-12-26 2004-11-04 Toomas Neuman Methods and compositions for the diagnosis, prognosis, and treatment of cancer
US20050112630A1 (en) * 2001-11-07 2005-05-26 Shaughnessy John D. Diagnosis, prognosis and identification of potential therapeutic targets of multiple myeloma based on gene expression profiling
US20050130117A1 (en) * 2001-10-03 2005-06-16 Davis Cong L. Modulators of lymphocyte activation and migration
US20050147593A1 (en) * 2003-05-22 2005-07-07 Medimmune, Inc. EphA2, EphA4 and LMW-PTP and methods of treatment of hyperproliferative cell disorders
US20050153425A1 (en) * 2002-12-20 2005-07-14 Xiao Xu Real time electronic cell sensing systems and applications for cell-based assays
US20050153923A1 (en) * 2003-12-04 2005-07-14 Kinch Michael S. Targeted drug delivery using EphA2 or EphA4 binding moieties
US20050245475A1 (en) * 2002-11-14 2005-11-03 Dharmacon, Inc. Functional and hyperfunctional siRNA directed against Bcl-2
US20050267059A1 (en) * 2003-11-14 2005-12-01 Diana Beardsley Syk-targeted nucleic acid interference
US20060003322A1 (en) * 2002-11-14 2006-01-05 Isaac Bentwich Bioinformatically detectable group of novel regulatory genes and uses thereof
US20060058255A1 (en) * 2004-03-01 2006-03-16 Jianzhu Chen RNAi-based therapeutics for allergic rhinitis and asthma
US20060073474A1 (en) * 2001-07-10 2006-04-06 Perez Omar D Methods and compositions for detecting the activation state of multiple proteins in single cells
US20060141493A1 (en) * 2001-11-09 2006-06-29 Duke University Office Of Science And Technology Atherosclerotic phenotype determinative genes and methods for using the same
US20060150262A1 (en) * 2003-02-05 2006-07-06 Jadwiga Bienkowska Novel mucin-like polypeptides

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989005358A1 (en) 1987-11-30 1989-06-15 University Of Iowa Research Foundation Dna and rna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes
US5256555A (en) 1991-12-20 1993-10-26 Ambion, Inc. Compositions and methods for increasing the yields of in vitro RNA transcription and other polynucleotide synthetic reactions
US5939262A (en) 1996-07-03 1999-08-17 Ambion, Inc. Ribonuclease resistant RNA preparation and utilization
US6057134A (en) 1996-10-07 2000-05-02 Ambion, Inc. Modulating the efficiency of nucleic acid amplification reactions with 3' modified oligonucleotides
US6111086A (en) 1998-02-27 2000-08-29 Scaringe; Stephen A. Orthoester protecting groups
AU2001245793A1 (en) * 2000-03-16 2001-09-24 Cold Spring Harbor Laboratory Methods and compositions for rna interference
US20030084471A1 (en) 2000-03-16 2003-05-01 David Beach Methods and compositions for RNA interference
AU2001249622B2 (en) 2000-03-30 2007-06-07 Massachusetts Institute Of Technology RNA sequence-specific mediators of RNA interference
ES2728168T3 (en) * 2000-12-01 2019-10-22 Max Planck Gesellschaft Small RNA molecules that mediate RNA interference
JP2004536800A (en) * 2001-05-09 2004-12-09 アルカベロ アクチェセルスカプ Pharmaceutical composition for preventing or treating TH1 and TH2 cell-related diseases by modulating the TH1 / TH2 ratio
US20060127385A1 (en) * 2002-12-06 2006-06-15 The Trustees Of Boston University Method for sustaining enos activity

Patent Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020068703A1 (en) * 1993-09-30 2002-06-06 University Of Pennsylvania. Methods of inhibiting phagocytosis
US6242427B1 (en) * 1993-09-30 2001-06-05 University Of Pennsylvania Methods of inhibiting phagocytosis
US20060073474A1 (en) * 2001-07-10 2006-04-06 Perez Omar D Methods and compositions for detecting the activation state of multiple proteins in single cells
US20050130117A1 (en) * 2001-10-03 2005-06-16 Davis Cong L. Modulators of lymphocyte activation and migration
US20050112630A1 (en) * 2001-11-07 2005-05-26 Shaughnessy John D. Diagnosis, prognosis and identification of potential therapeutic targets of multiple myeloma based on gene expression profiling
US20030232364A1 (en) * 2001-11-07 2003-12-18 Shaughnessy John D. Diagnosis, prognosis and identification of potential therapeutic targets of multiple myeloma based on gene expression profiling
US20040009523A1 (en) * 2001-11-07 2004-01-15 Shaughnessy John D. Diagnosis, prognosis and identification of potential therapeutic targets of multiple myeloma based on gene expression profiling
US20060141493A1 (en) * 2001-11-09 2006-06-29 Duke University Office Of Science And Technology Atherosclerotic phenotype determinative genes and methods for using the same
US20040018522A1 (en) * 2002-05-09 2004-01-29 Brigham And Women's Hospital, Inc. Identification of dysregulated genes in patients with multiple sclerosis
US20040028685A1 (en) * 2002-05-10 2004-02-12 Kinch Michael S. EphA2 monoclonal antibodies and methods of use thereof
US20040106156A1 (en) * 2002-07-10 2004-06-03 Perez Omar D. Methods and compositions for detecting receptor-ligand interactions in single cells
US20050245475A1 (en) * 2002-11-14 2005-11-03 Dharmacon, Inc. Functional and hyperfunctional siRNA directed against Bcl-2
US20050246794A1 (en) * 2002-11-14 2005-11-03 Dharmacon Inc. Functional and hyperfunctional siRNA
US20060003322A1 (en) * 2002-11-14 2006-01-05 Isaac Bentwich Bioinformatically detectable group of novel regulatory genes and uses thereof
US20050153425A1 (en) * 2002-12-20 2005-07-14 Xiao Xu Real time electronic cell sensing systems and applications for cell-based assays
US20040219575A1 (en) * 2002-12-26 2004-11-04 Toomas Neuman Methods and compositions for the diagnosis, prognosis, and treatment of cancer
US20060150262A1 (en) * 2003-02-05 2006-07-06 Jadwiga Bienkowska Novel mucin-like polypeptides
US20050147593A1 (en) * 2003-05-22 2005-07-07 Medimmune, Inc. EphA2, EphA4 and LMW-PTP and methods of treatment of hyperproliferative cell disorders
US20050267059A1 (en) * 2003-11-14 2005-12-01 Diana Beardsley Syk-targeted nucleic acid interference
US20050153923A1 (en) * 2003-12-04 2005-07-14 Kinch Michael S. Targeted drug delivery using EphA2 or EphA4 binding moieties
US20060058255A1 (en) * 2004-03-01 2006-03-16 Jianzhu Chen RNAi-based therapeutics for allergic rhinitis and asthma

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110124567A1 (en) * 2000-04-13 2011-05-26 Wight Thomas N Therapeutic compounds and methods
WO2009061368A1 (en) * 2007-11-06 2009-05-14 Benaroya Research Institute Inhibition of versican with sirna and other molecules
US20110008366A1 (en) * 2007-11-06 2011-01-13 Wight Thomas N Inhibition of versican with sirna and other molecules
US8410067B2 (en) 2007-11-06 2013-04-02 Benaroya Research Institute Inhibition of versican with siRNA and other molecules
WO2011100341A1 (en) * 2010-02-09 2011-08-18 Drexel University New signaling molecule involved in ultraviolet damage to skin
US9551710B2 (en) 2010-02-09 2017-01-24 Drexel University Signaling molecule involved in ultraviolet damage to skin
WO2014071109A1 (en) 2012-11-01 2014-05-08 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
WO2014151386A1 (en) 2013-03-15 2014-09-25 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
WO2014194254A1 (en) 2013-05-30 2014-12-04 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
EP3811974A1 (en) 2013-05-30 2021-04-28 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
WO2015051244A1 (en) 2013-10-04 2015-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015051241A1 (en) 2013-10-04 2015-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
EP3964507A1 (en) 2013-10-04 2022-03-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015061204A1 (en) 2013-10-21 2015-04-30 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2015143012A1 (en) 2014-03-19 2015-09-24 Infinity Pharmaceuticals, Inc. Heterocyclic compounds for use in the treatment of pi3k-gamma mediated disorders
EP4066834A1 (en) 2014-03-19 2022-10-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds for use in the treatment of pi3k-gamma mediated disorders
WO2015160986A2 (en) 2014-04-16 2015-10-22 Infinity Pharmaceuticals, Inc. Combination therapies
WO2016054491A1 (en) 2014-10-03 2016-04-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2017214269A1 (en) 2016-06-08 2017-12-14 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2017223422A1 (en) 2016-06-24 2017-12-28 Infinity Pharmaceuticals, Inc. Combination therapies

Also Published As

Publication number Publication date
JP2011200238A (en) 2011-10-13
WO2005007623A2 (en) 2005-01-27
AU2004257167A1 (en) 2005-01-27
WO2005007623A3 (en) 2006-05-18
EP1692153A2 (en) 2006-08-23
CN1860127A (en) 2006-11-08
US20050075306A1 (en) 2005-04-07
AU2004257167B2 (en) 2012-03-29
CN100577680C (en) 2010-01-06
HK1096972A1 (en) 2007-06-15
EP2371835A1 (en) 2011-10-05
EP1692153A4 (en) 2007-03-21
US20120093913A1 (en) 2012-04-19
CA2531069A1 (en) 2005-01-27
JP2007524397A (en) 2007-08-30
US7173015B2 (en) 2007-02-06
AU2004257167A2 (en) 2005-01-27
CN101961497A (en) 2011-02-02
WO2005007623A8 (en) 2006-03-23

Similar Documents

Publication Publication Date Title
US7173015B2 (en) Inhibition of Syk kinase expression
Choung et al. Chemical modification of siRNAs to improve serum stability without loss of efficacy
EP3236976B1 (en) Rna interference agents for p21 gene modulation
Leirdal et al. Gene silencing in mammalian cells by preformed small RNA duplexes
Lage MDR1/P-glycoprotein (ABCB1) as target for RNA interference-mediated reversal of multidrug resistance
Yokota et al. siRNA-based inhibition specific for mutant SOD1 with single nucleotide alternation in familial ALS, compared with ribozyme and DNA enzyme
US20100055783A1 (en) Nucleic acid compounds for inhibiting ras gene expression and uses thereof
Petrova et al. 2'-O-Methyl–Modified Anti-MDR1 Fork-siRNA Duplexes Exhibiting High Nuclease Resistance and Prolonged Silencing Activity
US8318924B2 (en) Immunostimulatory siRNA molecules
Chen et al. Downregulation of SOK1 promotes the migration of MCF-7 cells
US20080299659A1 (en) Nucleic acid compounds for inhibiting apob gene expression and uses thereof
EP1614751B1 (en) Oligoribonucleotide sequence homologous to a cdna region which codes for the human cd40 receptor and duplex oligoribonucleotides, vectors, pharmaceutical compositions and uses associated thereto
Holen et al. Lobotomy of genes: use of RNA interference in neuroscience
US20220298512A1 (en) Sirna sequences targeting the expression of human genes jak1 or jak3 for a therapeutic use
US9540644B2 (en) Small interference RNA for inhibiting intracellular expression of ribosomal protein S3
Yoo et al. Competition potency of siRNA is specified by the 5′-half sequence of the guide strand
Oridate et al. Suppression of DNA methyltransferase 1 levels in head and neck squamous carcinoma cells using small interfering RNA results in growth inhibition and increase in Cdk inhibitor p21
Wang et al. Ribozyme-and siRNA-mediated suppression of RGS-containing RhoGEF proteins
CA2690732A1 (en) Rnai mediated knockdown of numa for cancer therapy
Calderón et al. RNA interference: a novel and physiologic mechanism of gene silencing with great therapeutic potential
Ui-Tei et al. RNAi induced in mammalian and Drosophila cells via transfection of dimers and trimers of small interfering RNA
US20060189560A1 (en) Molecular method to augment RNA mediated gene silencing
EP1507004A1 (en) Method to inhibit the propagation of an undesired cell population
TW201718854A (en) RNA interference agents for p21 gene modulation

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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