WO2011130065A1 - RNA INTERFERENCE MEDIATED INHIBITION OF MET GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA) - Google Patents

RNA INTERFERENCE MEDIATED INHIBITION OF MET GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA) Download PDF

Info

Publication number
WO2011130065A1
WO2011130065A1 PCT/US2011/031247 US2011031247W WO2011130065A1 WO 2011130065 A1 WO2011130065 A1 WO 2011130065A1 US 2011031247 W US2011031247 W US 2011031247W WO 2011130065 A1 WO2011130065 A1 WO 2011130065A1
Authority
WO
WIPO (PCT)
Prior art keywords
sina
nucleotides
nucleic acid
double
short interfering
Prior art date
Application number
PCT/US2011/031247
Other languages
French (fr)
Inventor
Steve Bartz
Duncan Brown
Elizabeth Anne Harrington
Yong Ma
Ulrike Philippar
Original Assignee
Merck Sharp & Dohme Corp.
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 Merck Sharp & Dohme Corp. filed Critical Merck Sharp & Dohme Corp.
Publication of WO2011130065A1 publication Critical patent/WO2011130065A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • 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
    • C12N15/1135Non-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 oncogenes or tumor suppressor genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates

Definitions

  • sequence listing submitted via EFS in compliance with 37 CFR ⁇ 1.52(e)(5), is incorporated herein by reference.
  • the sequence listing text file submitted via EFS contains the file "SequenceListing_SIRONCl", created on April 4, 2011, which is 721,386 bytes in size.
  • RTK Receptor tyrosine kinases
  • MET a subfamily of receptor tyrosine kinases
  • RON receptors can function as oncogenes like most tyrosine kinases.
  • MET has been shown to be overexpressed and/or mutated in a variety of malignancies.
  • the MET proto-oncogene encodes the MET receptor tyrosine kinase.
  • the MET receptor is an approximately 190kDa glycosylated dimeric complex composed of a 50kDa alpha chain disulfide-linked to a 145kDa beta chain. The alpha chain is found extracellularly while the beta chain contains extracellular, transmembrane, and cytosolic domains.
  • the c-MET protein is synthesized as a precursor and is proteolytically cleaved to yield mature alpha and beta subunits. It displays structural similarities to semaphoring and plexins, a ligand-receptor family that is involved in cell-cell interaction. SIR-ONC-00001
  • HGF hepatocyte growth factor
  • HGF/SF hepatocyte growth factor
  • binding of HGF to the MET extracellular domain results in receptor multimerization and phosphorylation of multiple tyrosine residues in the MET intracellular domain.
  • Activation of MET signaling can lead to a wide array of cellular responses including proliferation, survival, angiogenesis, wound healing, tissue regeneration, scattering, motility, invasion and branching morphogenesis.
  • HGF/MET signaling also plays a major role in the invasive growth that is found in most tissues, including cartilage, bone, blood vessels, and neurons.
  • HCC hepatocellular carcinomas
  • MET and its ligand HGF have been implicated in progression of several types of human cancers including liver, prostate, colon, breast, and skin cancers.
  • MET mutations have been well described in multiple solid tumors and some hematologic malignancies.
  • the prototypic MET mutation examples are seen in hereditary and sporadic human papillary renal carcinoma (Schmidt, L. et al., Nat. Tenet. 1997, 16, 68-73; Jeffers, M. et al, Proc. Nat. Acad. Sci. 1997, 94, 11445-11500).
  • Other reported examples of MET mutations include ovarian cancer, childhood hepatocellular carcinoma, metastatic head and neck squamous cell carcinomas and gastric cancers.
  • HGF/MET has been shown to inhibit anoikis, suspension- induced programmed cell death (apoptosis), in head and neck squamous cell carcinoma cells.
  • MET signaling is implicated in various cancers, especially renal.
  • the nexus between MET and colorectal cancer has also been established.
  • Analysis of MET expression during colorectal cancer progression showed that 50% of the carcinoma specimens analyzed expressed 5 to 50-fold higher levels of MET mRNA transcripts and protein versus the adjacent normal colonic mucosa.
  • 70% of colorectal cancer liver metastasis showed MET overexpression.
  • MET is also implicated in glioblastoma.
  • High-grade malignant gliomas are the most common cancers of the central nervous system. Despite treatment with surgical resection, radiation therapy, and chemotherapy, the mean overall survival is ⁇ 1.5 years, and few patients survive for > 3 years.
  • Human malignant gliomas frequently express both HGF and MET, which can establish an autocrine loop of biological significance.
  • Glioma MET expression correlates with glioma grade, and an analysis of human tumor specimens showed that malignant gliomas have a 7-fold higher HGF content than low-grade gliomas.
  • Multiple studies have demonstrated that human gliomas frequently co-express HGF and MET and that high levels of expression are associated with malignant progression. It was further shown that HGF-MET is able to activate Akt and protect glioma cell lines from apoptotic death, both in vitro and in vivo.
  • RNAi RNA interference
  • siNA short interfering nucleic acids
  • siRNA messenger RNAs
  • RISC RNA-induced silencing complex
  • RNA interference can also involve small RNA ⁇ e.g., micro-RNA or miRNA) mediated gene silencing, presumably though cellular mechanisms that either inhibit translation or that regulate chromatin structure and thereby prevent transcription of target gene sequences (see for example Allshire, 2002, Science, 29 ' , 1818-1819; Volpe et ah, 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al, 2002, Science, 297, 2232-2237).
  • RNAi small RNA ⁇ e.g., micro-RNA or miRNA
  • the invention provides a solution to the problem of treating diseases that respond to the modulation of the MET gene expression using novel short interfering nucleic acid (siNA) molecules to modulate MET expression.
  • siNA short interfering nucleic acid
  • the present invention provides compounds, compositions, and methods useful for modulating the expression of MET genes, specifically those MET genes associated with cancer and for treating such conditions by RNA interference (RNAi) using small nucleic acid molecules.
  • RNAi RNA interference
  • the instant invention features small nucleic acid molecules, i.e., short interfering nucleic acid (siNA) molecules including, but not limited to, short interfering RNA (siRNA), double-stranded RNA (dsRNA), micro-RNA (miRNA), short hairpin RNA (shRNA) and circular RNA molecules and methods used to modulate the expression of MET genes and/or other genes involved in pathways of MET gene expression and/or activity.
  • siNA short interfering nucleic acid
  • the invention provides double-stranded short interfering nucleic acid (siNA) molecules that inhibit the expression of a MET gene in a cell or mammal, wherein the double-stranded siNAs comprise a sense and an antisense stand.
  • the antisense strand comprises a sequence that is complementary to at least a part of an RNA associated with the expression of the MET gene.
  • the sense strand comprises a sequence that is complementary to the antisense strand.
  • the antisense strand comprises at least 15 nucleotides having sequence complementarity to a target sequence set forth in Table la.
  • the antisense strand comprises at least a 15 nucleotide sequence of one of the antisense sequences set forth in Table lb.
  • the sense strand comprises at least a 15 nucleotide sequence of a sense strand sequence as set forth in Table lb.
  • double-stranded short interfering nucleic acid (siNA) molecules wherein the antisense stand comprises a modified sequence as set forth in Table lc that has sequence complementarity to a target sequence of the invention.
  • the sense strand also comprises a modified sequence as set forth in Table lc. SIR-ONC-00001
  • the present invention provides a double- stranded short interfering nucleic acid (siNA) molecule that modulates the expression of MET, wherein the siNA comprises a sense strand and an antisense strand; each strand is independently 15 to 30 nucleotides in length; and the antisense strand comprises at least 15 nucleotides having sequence complementary to any of:
  • the antisense strand of a siNA molecule comprises at least a 15 nucleotide sequence of:
  • the sense strand of a siNA molecule of the invention comprises at least a 15 nucleotide sequence of:
  • a siNA molecule of the invention comprises any of:
  • nucleotides of siNAs of the invention are unmodified.
  • one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) of the nucleotide positions independently in either one or both strands of an siNA molecule are modified.
  • Modifications include nucleic acid sugar modifications, base modifications, backbone (internucleotide linkage) modifications, non-nucleotide modifications, and/or any combination thereof.
  • purine and pyrimidine nucleotides are differentially modified.
  • purine and pyrimidine nucleotides can be differentially modified at the 2'- sugar position (i.e., at least one purine has a different modification from at least one pyrimidine in the same or different strand at the 2'-sugar position).
  • the purines are unmodified in one or both strands, while the pyrimidines in one or both strands are modified.
  • the pyrimidines are unmodified in one or both strands, while the purines in one or both strands are modified.
  • At least one modified nucleotide is a 2'-deoxy-2'-fluoro nucleotide, a 2'-deoxy nucleotide, or a 2'-0-alkyl nucleotide.
  • at least 5 or more of the pyrimidine nucleotides in one or both stands are either all 2'-deoxy-2'-fluoro or all 2'-0-methyl pyrimidine nucleotides.
  • at least 5 or more of the purine nucleotides in one or both stands are either all 2'-deoxy-2'-fluoro or all 2'-0-methyl purine nucleotides.
  • the siNA molecules comprise one or more modifications as described herein, the nucleotides at positions 1, 2, and 3 at the 5' end of the guide (antisense) strand are unmodified.
  • the siNA molecules of the invention have 3' overhangs of one, two, three, or four nucleotide(s) on one or both of the strands.
  • the siNA molecules lack overhangs (i.e., have blunt ends).
  • the siNA molecule has 3' overhangs of two nucleotides on both the sense and antisense strands. The overhangs can be SIR-ONC-00001 modified or unmodified.
  • modified nucleotides in the overhangs include, but are not limited to, 2'-0-alkyl nucleotides, 2'-deoxy-2'-fluoro nucleotides, locked nucleic acid (LNA) nucleotides, or 2'-deoxy nucleotides.
  • the overhang nucleotides in the antisense strand can comprise nucleotides that are complementary to nucleotides in the MET target sequence.
  • the overhangs in the sense stand can comprise nucleotides that are in the MET target sequence.
  • the siNA molecules of the invention have two 3' overhang nucleotides on the antisense stand that are 2'-0-alkyl (e.g., 2'-0-methyl) nucleotides and two 3' overhang nucleotides on the sense stand that are 2'-deoxy nucleotides.
  • the siNA molecules of the invention have two 3' overhang nucleotides that are 2'-0-alkyl (e.g., 2'-0- methyl) nucleotides on both the antisense stand and on the sense stand.
  • the 2'-0-alkyl nucleotides are 2'-0-methyl uridine nucleotides.
  • the overhangs also comprise one or more phosphorothioate linkages between nucleotides of the overhang.
  • the siNA molecules of the invention have caps (also referred to herein as "terminal caps.”
  • the cap can be present at the 5'-terminus (5'-cap) or at the 3'- terminus (3'-cap) or can be present on both termini, such as at the 5' and 3' termini of the sense strand of the siNA.
  • the siNA molecules of the invention when double stranded can be symmetric or asymmetric.
  • Each strand of these double stranded siNAs independently can range in nucleotide length between 3 and 30 nucleotides.
  • each strand of the siNA molecules of the invention is about 15 to 30 (i.e., about 19, 20, 21, 22, 23 or 24) nucleotides in length.
  • the siNA molecules of the invention which are double stranded or have a duplex structure, independently comprise about 3 to about 30 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) base pairs.
  • the duplex structure of siNAs of the invention is between 15 and 30, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 base pairs in length.
  • double-stranded short interfering nucleic acid (siNA) molecules are provided, wherein the molecule has a sense strand and an antisense strand and comprises formula (A): SIR-ONC-00001
  • the upper strand is the sense strand and the lower strand is the antisense strand of the double- stranded nucleic acid molecule; wherein the antisense strand comprises at least a 15 nucleotide sequence of SEQ ID NO: 1049, SEQ ID NO: 1091, SEQ ID NO: 1099, or SEQ ID NO: 1101, and the sense strand comprises a sequence having complementarity to the antisense strand; each N is independently a nucleotide which is unmodified or chemically modified or a non-nucleotide; each B is a terminal cap that is present or absent;
  • (N) represents overhanging nucleotides, each of which is independently unmodified or chemically modified
  • [N] represents nucleotides that are ribonucleotides
  • XI and X2 are independently integers from 0 to 4
  • X3 is an integer from 15 to 30
  • X4 is an integer from 9 to 30;
  • X5 is an integer from 0 to 6, provided that the sum of X4 and X5 is 15-30.
  • the invention features a double- stranded short interfering nucleic acid (siNA) of formula (A); wherein
  • one or more pyrimidine nucleotides in ⁇ 4 positions are independently 2'-deoxy- 2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof; SIR-ONC-00001
  • one or more purine nucleotides in ⁇ 4 positions are independently 2'-deoxy-2'- fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof;
  • one or more pyrimidine nucleotides in ⁇ 3 positions are independently 2'-deoxy- 2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof;
  • one or more purine nucleotides in ⁇ 3 positions are independently 2'-deoxy-2'- fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides,
  • compositions comprising the double-stranded nucleic acids molecules described herein with optionally a pharmaceutically acceptable carrier or diluent.
  • composition can be carried out by known methods, wherein the nucleic acid is introduced into a desired target cell in vitro or in vivo.
  • nucleic acid molecules of the invention include the use of various carrier systems, reagents and vectors.
  • carrier systems suitable for use in the present invention include conjugates, nucleic-acid-lipid particles, lipid nanoparticles (LNP), liposomes, lipoplexes, micelles, virosomes, virus like particles (VLP), nucleic acid complexes, and mixtures thereof.
  • LNP lipid nanoparticles
  • VLP virus like particles
  • compositions of the invention can be in the form of an aerosol, dispersion, solution (e.g., an injectable solution), a cream, ointment, tablet, powder, suspension or the like. These compositions may be administered in any suitable way, e.g. orally, sublingually, buccally, parenterally, nasally, or topically. In some embodiments, the compositions are aerosolized and delivered via inhalation.
  • the molecules and compositions of the present invention have utility over a broad range of therapeutic applications. Accordingly another aspect of this invention relates to the use of the compounds and compositions of the invention in treating a subject.
  • the invention thus provides a method for treating a subject, such as a human, suffering from a condition which is mediated by the action, or by the loss of action, of MET, wherein the method comprises SIR-ONC-00001 administering to the subject an effective amount of a double-stranded short interfering nucleic acid (siNA) molecule of the invention.
  • the condition is cancer.
  • Figure 1 shows a non-limiting proposed mechanistic representation of target RNA degradation involved in RNAi.
  • Double-stranded RNA dsRNA
  • RdRP RNA-dependent RNA polymerase
  • siNA duplexes RNA-dependent RNA polymerase
  • synthetic or expressed siNA can be introduced directly into a cell by appropriate means.
  • An active siNA complex forms that recognizes a target RNA, resulting in degradation of the target RNA by the RISC endonuclease complex or in the synthesis of additional RNA by RNA-dependent RNA polymerase (RdRP), which can activate DICER and result in additional siNA molecules, thereby amplifying the RNAi response.
  • RdRP RNA-dependent RNA polymerase
  • Figure 2 shows non-limiting examples of chemically modified siNA constructs of the present invention using a generalized structure of a representative siNA duplex.
  • the specific modifications shown in the figure can be utilized alone or in combination with other modifications of the figure, in addition to other modifications and features described herein with reference to any siNA molecule of the invention.
  • N stands for any nucleotide or optionally a non-nucleotide as described here.
  • the upper strand, having ⁇ - ⁇ 3 -( ⁇ ) ⁇ 2 - ⁇ -3' is the sense (or passenger) strand of the siNA
  • the lower strand, having B(N)xi-Nx 4 -[N]x5 -5' is the antisense (or guide) strand of the siNA.
  • Nucleotides (or optional non-nucleotides) of internal portions of the sense strand are designated Nxs and nucleotides (or optional non- nucleotides) of internal portions of the antisense strand are designated ⁇ 4 .
  • Nucleotides (or optional non-nucleotides) of the internal portions are generally base paired between the two strands, but can optionally lack base pairing (e.g.
  • Nucleotides (or optional non-nucleotides) of overhang regions are designated by parenthesis (N).
  • Nucleotides of the 5'-terminal portion of the antisense strand are designated [N].
  • Terminal caps are optionally present at the 5' and/or 3' end of the sense strand and further optionally present at the 3'-end of the antisense strand.
  • each strand can independently range from about 15 to about 30 nucleotides in length, but can vary depending on the presence of any overhang nucleotides.
  • XI and X2 are independently integers from 0 to 4; X3 is an integer from 15 to 30; X4 is an integer from 9 to 30; X5 is an integer from 0 to 6, provided that the sum of X4 and X5 is 15-30.
  • Various modifications are shown for the nucleotides of the sense and antisense strands of the siNA constructs.
  • the (N) overhang nucleotide positions can be chemically modified as described herein (e.g., 2'-0-methyl, 2'- deoxy-2'-fluoro, 2'-deoxy, LNA, universal bases etc.) and can be either derived from a corresponding target nucleic acid sequence or not.
  • the constructs shown in the figure can also comprise phosphorothioate linkages as described herein.
  • phosphorothioate linkages can exist between any N, (N), and/or [N] positions.
  • Such phosphorothioate incorporation can be utilized between purine "R” and pyrimidine “Y” positions, or for stabilization of pyrimidine linkages in general.
  • the constructs shown in the figure can optionally include a ribonucleotide at the 9 th position from the 5'-end of the sense strand or the 11 th position based on the 5'-end of the guide strand by counting 11 nucleotide positions in from the 5'-terminus of the guide strand.
  • the antisense strand can include a ribonucleotide at the 14 th position from the 5'-end, or alternately can be selected or designed so that a 2'-0-alkyl nucleotide (e.g., a 2'-0-methyl purine) is not present at this position.
  • a 2'-0-alkyl nucleotide e.g., a 2'-0-methyl purine
  • the 5'-terminal position of the antisense strand can comprise a terminal phosphate group as described herein.
  • the antisense strand generally comprises sequence complementary to any target nucleic acid sequence of the invention, such as those set forth in Table la herein.
  • Figure 3 shows non-limiting examples of certain combinations of modifications applied to the representative siNA duplex described in Figure 2.
  • the table shown below the representative structure provides specific combinations of ( ⁇ ) ⁇ 1; ( ⁇ ) ⁇ 2 , ⁇ 3, ⁇ 4 , and/or [ ⁇ ] ⁇ 5 nucleotide (and optional non-nucleotide) positions.
  • combinations of 5 or more (e.g., 5, 6, 7, 8, 9, or 10 or more) ⁇ 3 and 5 or more (e.g., 5, 6, 7, 8, 9, or 10 or more) ⁇ 4 pyrimidine "Y" and purine "R" nucleotides are specified, each of which can independently have SIR-ONC-00001 specific ( ⁇ ) ⁇ 1 ; and/or ( ⁇ ) ⁇ 2 , substitutions as shown in the figure, in addition to optional phosphorothioate substitutions.
  • the 5'-terminal antisense strand [N] nucleotides are generally ribonucleotides, but can also be modified or unmodified depending on if they are purine "R" or pyrimidine "Y" nucleotides
  • Figure 4A-C shows non-limiting examples of different siNA constructs of the invention.
  • the criteria of the representative structures shown in Figures 2 and 3 can be applied to any of the structures shown in Figure 4A-C.
  • constructs 1, 2, and 3 have 19 representative base pairs; however, different embodiments of the invention include any number of base pairs described herein. Bracketed regions represent nucleotide overhangs, for example, comprising about 1, 2, 3, or 4 nucleotides in length, preferably about 2 nucleotides.
  • Constructs 1 and 2 can be used independently for RNAi activity.
  • Construct 2 can comprise a polynucleotide or non- nucleotide linker, which can optionally be designed as a biodegradable linker.
  • the loop structure shown in construct 2 can comprise a biodegradable linker that results in the formation of construct 1 in vivo and/or in vitro.
  • construct 3 can be used to generate construct 2 under the same principle wherein a linker is used to generate the active siNA construct 2 in vivo and/or in vitro, which can optionally utilize another biodegradable linker to generate the active siNA construct 1 in vivo and/or in vitro.
  • a linker is used to generate the active siNA construct 2 in vivo and/or in vitro, which can optionally utilize another biodegradable linker to generate the active siNA construct 1 in vivo and/or in vitro.
  • the stability and/or activity of the siNA constructs can be modulated based on the design of the siNA construct for use in vivo or in vitro and/or in vitro.
  • the examples shown in Figure 4B represent different variations of double-stranded nucleic acid molecule of the invention, such as microRNA, that can include overhangs, bulges, loops, and stem-loops resulting from partial complementarity.
  • Such motifs having bulges, loops, and stem-loops are generally characteristics of miRNA.
  • the bulges, loops, and stem-loops can result from any degree of partial complementarity, such as mismatches or bulges of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more nucleotides in one or both strands of the double-stranded nucleic acid molecule of the invention.
  • the example shown in Figure 4C represents a model double- stranded nucleic acid molecule of the invention comprising a 19 base pair duplex of two 21 nucleotide sequences SIR-ONC-00001 having dinucleotide 3 '-overhangs.
  • the top strand (1) represents the sense strand (passenger strand)
  • the middle strand (2) represents the antisense (guide strand)
  • the lower strand (3) represents a target polynucleotide sequence.
  • the dinucleotide overhangs (NN) can comprise a sequence derived from the target polynucleotide.
  • the 3'-(NN) sequence in the guide strand can be complementary to the 5'-[NN] sequence of the target polynucleotide.
  • the 5'-(NN) sequence of the passenger strand can comprise the same sequence as the 5'-[NN] sequence of the target polynucleotide sequence.
  • the overhangs (NN) are not derived from the target polynucleotide sequence, for example where the 3'-(NN) sequence in the guide strand are not complementary to the 5'-[NN] sequence of the target polynucleotide and the 5'-(NN) sequence of the passenger strand can comprise different sequence from the 5'-[NN] sequence of the target polynucleotide sequence.
  • any (NN) nucleotides are chemically modified, e.g., as 2'-0-methyl, 2'-deoxy-2'- fluoro, and/or other modifications herein.
  • the passenger strand can comprise a ribonucleotide position N of the passenger strand.
  • position N can be 9 nucleotides in from the 3' end of the passenger strand.
  • the position N is determined based on the 5'-end of the guide strand by counting 11 nucleotide positions in from the 5'-terminus of the guide strand and picking the corresponding base paired nucleotide in the passenger strand.
  • Cleavage by Ago2 takes place between positions 10 and 11 as indicated by the arrow.
  • Figure 6 shows a non-limiting example of a strategy used to identify chemically modified siNA constructs of the invention that are nuclease resistant while preserving the ability to mediate RNAi activity.
  • Chemical modifications are introduced into the siNA construct based on educated design parameters (e.g. introducing 2'-modifications, base modifications, backbone modifications, terminal cap modifications etc).
  • the modified construct is tested in an appropriate system (e.g., human serum for nuclease resistance, shown, or an animal model for PK/delivery parameters).
  • the siNA construct is tested for RNAi activity, for example in a cell culture system such as a luciferase reporter assay and/or against endogenous mRNA).
  • siNA constructs are then identified which possess a particular characteristic while maintaining RNAi activity, and can be further modified and assayed once again. This same approach can be used to identify siNA-conjugate molecules with improved pharmacokinetic profiles, delivery, and RNAi activity.
  • Figure 7 shows non-limiting examples of phosphorylated siNA molecules of the invention, including linear and duplex constructs and asymmetric derivatives thereof.
  • Figure 8 shows non-limiting examples of chemically modified terminal phosphate groups of the invention.
  • Figure 9 shows a non-limiting example of a cholesterol linked phosphoramidite that can be used to synthesize cholesterol conjugated siNA molecules of the invention.
  • An example is shown with the cholesterol moiety linked to the 5' -end of the sense strand of an siNA molecule.
  • Figure 10 depicts an embodiment of 5' and 3' inverted abasic cap linked to a nucleic acid strand.
  • Figure 11 shows the efficacy of various MET siNAs in an LNP formulation (DLinDMA/Cholesterol/S-PEG-C-DMA/DSPC in a 40/48/2/10 ratio with an N/P ratio of 6) at 21 days using a dosing schedule of the 6mpk/week in TRE-MET mice.
  • Figure 12 shows the log2(fold change) in MET mRNA gene expression (reported as AACt) in Rhesus macaque monkeys at 3, 7, 14 and 28 days post-dosing with various MET siNAs in an LNP formulation (DLinDMA/Cholesterol/S-PEG-C-DMA/DSPC in a 40/48/2/10 ratio with SIR-ONC-00001 an N/P ratio of 6).
  • the level of MET gene expression was also determined at 7 days pre-dosing.
  • PBS and an ApoB siNA were used as controls.
  • abasic refers to its meaning as is generally accepted in the art.
  • the term generally refers to sugar moieties lacking a nucleobase or having a hydrogen atom (H) or other non-nucleobase chemical groups in place of a nucleobase at the ⁇ position of the sugar moiety, see for example Adamic et ah, U.S. Pat. No. 5,998,203.
  • an abasic moiety of the invention is a ribose, deoxyribose, or dideoxyribose sugar.
  • acyclic nucleotide refers to its meaning as is generally accepted in the art.
  • the term generally refers to any nucleotide having an acyclic ribose sugar, for example where any of the ribose carbon/carbon or carbon/oxygen bonds are independently or in combination absent from the nucleotide.
  • alkyl refers to its meaning as is generally accepted in the art.
  • the term generally refers to a saturated or unsaturated hydrocarbons, including straight- chain, branched-chain, alkenyl, alkynyl groups and cyclic groups, but excludes aromatic groups. Notwithstanding the foregoing, alkyl also refers to non-aromatic heterocyclic groups.
  • the alkyl group has 1 to 12 carbons. More preferably, it is a lower alkyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
  • the alkyl group can be substituted or unsubstituted.
  • agents that interfere with cell cycle checkpoints refers to compounds that inhibit protein kinases that transduce cell cycle checkpoint signals, thereby sensitizing the cancer cell to DNA damaging agents.
  • RTKs receptor tyrosine kinases
  • androgen receptor modulators refers to compounds that interfere or inhibit the binding of androgens to the receptor, regardless of mechanism.
  • angiogenesis inhibitors refers to compounds that inhibit the formation of new blood vessels, regardless of mechanism.
  • aryl refers to its meaning as is generally accepted in the art.
  • the term generally refers to an aromatic group that has at least one ring having a conjugated pi electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which can be optionally substituted.
  • the preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, C1-C4 alkoxy, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, NH 2 , and NRiR 2 groups, where R and R 2 independently are H or C1-C4 alkyl.
  • alkylaryl refers to its meaning as is generally accepted in the art.
  • the term generally refers to an alkyl group (as described above) covalently joined to an aryl group (as described above).
  • Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted.
  • Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms.
  • Suitable heteroatoms include oxygen, sulfur, and nitrogen, and examples of heterocyclic aryl groups having such heteroatoms include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted.
  • the alkyl group is a C1-C4 alkyl group.
  • amide refers to its meaning as is generally accepted in the art.
  • the term generally refers to an -C(0)-NH-R, where R is either alkyl, aryl, alkylaryl or hydrogen.
  • the phrase "antisense region” as used herein refers to its meaning as is generally accepted in the art.
  • the SIR-ONC-00001 term refers to a nucleotide sequence of an siNA molecule having complementarity to a target nucleic acid sequence.
  • the antisense region of an siNA molecule can optionally comprise a nucleic acid sequence having complementarity to a sense region of the siNA molecule.
  • the antisense region of the siNA molecule is referred to as the antisense strand or guide strand.
  • asymmetric hairpin refers to a linear siNA molecule comprising an antisense region, a loop portion that can comprise nucleotides or non-nucleotides, and a sense region that comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complementary nucleotides to base pair with the antisense region and form a duplex with loop.
  • an asymmetric hairpin siNA molecule of the invention can comprise an antisense region having length sufficient to mediate RNAi in a cell or in vitro system ⁇ e.g.
  • the asymmetric hairpin siNA molecule can also comprise a 5 '-terminal phosphate group that can be chemically modified.
  • the loop portion of the asymmetric hairpin siNA molecule can comprise nucleotides, non-nucleotides, linker molecules, or conjugate molecules as described herein.
  • biodegradable refers to its meaning as is generally accepted in the art.
  • the term generally refers to degradation in a biological system, for example, enzymatic degradation or chemical degradation.
  • biodegradable linker refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a linker molecule that is designed to connect one molecule to another molecule, and which is susceptible to degradation in a biological system.
  • the linker can be a nucleic acid or non-nucleic acid based linker.
  • a biodegradable linker can be used to attach a ligand or biologically active molecule to an siNA molecule of the invention. Alternately, a biodegradable linker can be used to connect the sense and antisense strands of an siNA molecule SIR-ONC-00001 of the invention.
  • the biodegradable linker is designed such that its stability can be modulated for a particular purpose, such as delivery to a particular tissue or cell type.
  • the stability of a nucleic acid-based biodegradable linker molecule can be modulated by using various chemistries, for example combinations of ribonucleotides, deoxyribonucleotides, and chemically modified nucleotides, such as 2'-0-methyl, 2'-fluoro, 2'-amino, 2'-0-amino, 2'-C-allyl, 2'-0-allyl, and other 2'-modified or base modified nucleotides.
  • the biodegradable nucleic acid linker molecule can be a dimer, trimer, tetramer or longer nucleic acid molecule, for example, an oligonucleotide of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides in length, or can comprise a single nucleotide with a phosphorus-based linkage, for example, a phosphoramidate or phosphodiester linkage.
  • the biodegradable nucleic acid linker molecule can also comprise nucleic acid backbone, nucleic acid sugar, or nucleic acid base modifications.
  • biologically active molecule refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to compounds or molecules that are capable of eliciting or modifying a biological response in a system and/or are capable of modulating the pharmacokinetics and/or pharmacodynamics of other biologically active molecules.
  • biologically active molecules include siNA molecules alone or in combination with other molecules including, but not limited to therapeutically active molecules such as antibodies, cholesterol, hormones, antivirals, peptides, proteins, chemotherapeutics, small molecules, vitamins, co-factors, nucleosides, nucleotides, oligonucleotides, enzymatic nucleic acids, antisense nucleic acids, triplex forming oligonucleotides, polyamines, polyamides, polyethylene glycol, other polyethers, 2-5A chimeras, siNA, dsRNA, allozymes, aptamers, decoys and analogs thereof.
  • therapeutically active molecules such as antibodies, cholesterol, hormones, antivirals, peptides, proteins, chemotherapeutics, small molecules, vitamins, co-factors, nucleosides, nucleotides, oligonucleotides, enzymatic nucleic acids, antisense nucleic acids, triplex forming oligon
  • biological system refers to its meaning as is generally accepted in the art.
  • the term generally refers to material, in a purified or unpurified form, from biological sources including, but not limited to, human or animal, wherein the system comprises the components required for RNAi activity.
  • the phrase includes, for example, a cell, tissue, subject, or organism, or extract thereof.
  • the term also includes reconstituted material from a biological source.
  • blunt end refers to its meaning as is generally accepted in the art.
  • the term refers to termini of a double-stranded siNA molecule having no overhanging nucleotides.
  • the two strands of a double- stranded siNA molecule having blunt ends align with each other with matched base-pairs without overhanging nucleotides at the termini.
  • a siNA duplex molecule of the invention can comprise blunt ends at one or both termini of the duplex, such as termini located at the 5'-end of the antisense strand, the 5'-end of the sense strand, or both termini of the duplex.
  • cap also referred to herein as “terminal cap,” as used herein refers to its meaning as is generally accepted in the art.
  • the term refers to a moiety, which can be a chemically modified nucleotide or non-nucleotide that can be incorporated at one or more termini of one or more nucleic acid molecules of the invention. These terminal modifications protect the nucleic acid molecule from exonuclease degradation, and can help in delivery and/or localization within a cell.
  • the cap can be present at the 5'-terminus (5'-cap) or at the 3'-terminal (3'-cap) or can be present on both termini of any nucleic acid molecule of the invention.
  • a cap can be present at the 5'-end, 3-end and/or 5' and 3'-ends of the sense strand of a nucleic acid molecule of the invention. Additionally, a cap can optionally be present at the 3'-end of the antisense strand of a nucleic acid molecule of the invention.
  • the 5'-cap includes, but is not limited to, LNA; glyceryl; inverted deoxy abasic residue (moiety); 4',5'-methylene nucleotide; l-(beta- D-erythrofuranosyl) nucleotide, 4'-thio nucleotide; carbocyclic nucleotide; 1,5-anhydrohexitol nucleotide; L-nucleotides; alpha-nucleotides; modified base nucleotide; phosphorodithioate linkage; ⁇ /ireopentofuranosyl nucleotide; acyclic 3',4'-seco nucleotide; acyclic 3,4- dihydroxybutyl nucleotide; acyclic 3,5-dihydroxypentyl nucleotide; 3'-3'-inverted nucleotide moiety; 3'-3'-inverted abasic moiety;
  • Non-limiting examples of the 3'-cap include, but are not limited to, LNA; glyceryl; inverted deoxy abasic residue (moiety); 4', 5'-methylene nucleotide; l-(beta-D- erythrofuranosyl) nucleotide; 4'-thio nucleotide; carbocyclic nucleotide; 5'-amino-alkyl phosphate; l,3-diamino-2-propyl phosphate; 3 -aminopropyl phosphate; 6-aminohexyl phosphate; SIR-ONC-00001
  • the term "cell” as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term is used in its usual biological sense, and does not refer to an entire multicellular organism, e.g., specifically does not refer to a human being.
  • the cell can be present in an organism, e.g., birds, plants and mammals, such as humans, cows, sheep, apes, monkeys, swine, dogs, and cats.
  • the cell can be prokaryotic ⁇ e.g., bacterial cell) or eukaryotic ⁇ e.g., mammalian or plant cell).
  • the cell can be of somatic or germ line origin, totipotent or pluripotent, dividing or non-dividing.
  • the cell can also be derived from or can comprise a gamete or embryo, a stem cell, or a fully differentiated cell.
  • chemical modification refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to any modification of the chemical structure of the nucleotides that differs from nucleotides of native siRNA or RNA in general.
  • the term “chemical modification” encompasses the addition, substitution, or modification of native siRNA or RNA at the sugar, base, or internucleotide linkage, as described herein or as is otherwise known in the art.
  • the term “chemical modification” can refer to certain forms of RNA that are naturally occurring in certain biological systems, for example 2'-0-methyl modifications or inosine modifications.
  • MET refers to mesenchymal-epithelial transition factor which is a proto- oncogene that encodes MET proteins, MET peptides, MET polypeptides, MET regulatory polynucleotides (e.g., MET miRNAs and siNAs), mutant MET genes, and splice variants of a SIR-ONC-00001
  • MET genes as well as other genes involved in MET pathways of gene expression and/or activity. It is also known as the hepatocyte growth factor receptor (HGFR).
  • HGFR hepatocyte growth factor receptor
  • nucleic acid molecules of the invention refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the terms generally refer to the formation or existence of hydrogen bond(s) between one nucleic acid sequence and another nucleic acid sequence by either traditional Watson-Crick or other non-traditional types of bonding as described herein. In reference to the nucleic molecules of the present invention, the binding free energy for a nucleic acid molecule with its complementary sequence is sufficient to allow the relevant function of the nucleic acid to proceed, e.g., RNAi activity.
  • Partial complementarity can include various mismatches or non-based paired nucleotides (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more mismatches, non-nucleotide linkers, or non-based paired nucleotides) within the nucleic acid molecule, which can result in bulges, loops, or overhangs that result between the sense strand or sense region and the antisense strand or antisense region of the nucleic acid molecule or between the antisense strand or antisense region of the nucleic acid molecule and a corresponding target nucleic acid molecule.
  • mismatches or non-based paired nucleotides e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more mismatches, non-nucleotide linkers, or non-based paired nucleotides
  • Such partial complementarity can be represented by a % complementarity that is determined by the number of non-base paired nucleotides, i.e., about 50%, 60%, 70%, 80%, 90% etc. depending on the total number of nucleotides involved.
  • Such partial complementarity is permitted to the extent that the nucleic acid molecule (e.g., siNA) maintains its function, for example the ability to mediate sequence specific RNAi.
  • compositions or “formulation” as used herein refer to their generally accepted meaning in the art. These terms generally refer to a composition or formulation, such as in a pharmaceutically acceptable carrier or diluent, in a form suitable for administration, e.g., systemic or local administration, into a cell or subject, including, for example, a human. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, inhalation, or by injection. Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged nucleic acid is desirable for delivery). For example, compositions injected into the blood stream should be soluble.
  • compositions include formulations for human and veterinary use.
  • agents suitable for formulation with the nucleic acid molecules of the instant invention include: Lipid Nanoparticles (see for example Semple et al., 2010, Nat Biotechnol., Feb;28(2): 172-6.); P- glycoprotein inhibitors (such as Pluronic P85); biodegradable polymers, such as poly (DL- lactide-coglycolide) microspheres for sustained release delivery (Emerich, DF et al, 1999, Cell Transplant, 8, 47-58); and loaded nanoparticles, such as those made of polybutylcyanoacrylate.
  • a "pharmaceutically acceptable composition” or “pharmaceutically acceptable formulation” can refer to a composition or formulation that allows for the effective distribution of the nucleic acid molecules of the instant invention to the physical location most suitable for their desired activity.
  • cytotoxic/cytostatic agents refer to compounds that cause cell death or inhibit cell proliferation primarily by interfering directly with the cell's functioning or inhibit or interfere with cell mytosis, including alkylating agents, tumor necrosis factors, intercalators, hypoxia activatable compounds, microtubule inhibitors/micro tubule- stabilizing agents, inhibitors of mitotic kinesins, inhibitors of histone deacetylase, inhibitors of kinases involved in mitotic progression, antimetabolites; biological response modifiers; hormonal/anti-hormonal therapeutic SIR-ONC-00001 agents, hematopoietic growth factors, monoclonal antibody targeted therapeutic agents, topoisomerase inhibitors, proteasome inhibitors and ubiquitin ligase inhibitors.
  • estrogen receptor modulators refers to compounds that interfere with or inhibit the binding of estrogen to the receptor, regardless of mechanism.
  • gene or "target gene” as used herein refers to their meaning as is generally accepted in the art.
  • the terms generally refer a nucleic acid (e.g., DNA or RNA) sequence that comprises partial length or entire length coding sequences necessary for the production of a polypeptide.
  • the target gene can also include the UTR or non-coding region of the nucleic acid sequence.
  • a gene or target gene can also encode a functional RNA (fRNA) or non-coding RNA (ncRNA), such as small temporal RNA (stRNA), micro RNA (miRNA), small nuclear RNA (snRNA), short interfering RNA (siRNA), small nucleolar RNA (snRNA), ribosomal RNA (rRNA), transfer RNA (tRNA) and precursor RNAs thereof.
  • fRNA functional RNA
  • ncRNA non-coding RNA
  • stRNA small temporal RNA
  • miRNA micro RNA
  • snRNA small nuclear RNA
  • siRNA small nucleolar RNA
  • rRNA ribosomal RNA
  • tRNA transfer RNA
  • siNA molecules targeting fRNA and ncRNA can also be used to manipulate or alter the genotype or phenotype of a subject, organism or cell, by intervening in cellular processes such as genetic imprinting, transcription, translation, or nucleic acid processing (e.g., transamination, methylation etc.).
  • the target gene can be a gene derived from a cell, an endogenous gene, a transgene, or exogenous genes such as genes of a pathogen, for example a virus, which is present in the cell after infection thereof.
  • the cell containing the target gene can be derived from or contained in any organism, for example a plant, animal, protozoan, virus, bacterium, or fungus.
  • Non-limiting examples of plants include monocots, dicots, or gymnosperms.
  • Non-limiting examples of animals include vertebrates or invertebrates.
  • Non-limiting examples of fungi include molds or yeasts.
  • HMG-CoA reductase inhibitors refers to inhibitors of 3-hydroxy-3- methylglutaryl-CoA reductase.
  • HMG-CoA reductase inhibitor as used herein includes all pharmaceutically acceptable lactone and open-acid forms (i.e., where the lactone ring is SIR-ONC-00001 opened to form the free acid) as well as salt and ester forms of compounds that have HMG-CoA reductase inhibitory activity, and therefore the use of such salts, esters, open-acid and lactone forms is included within the scope of this invention.
  • homologous sequence refers to its meaning as is generally accepted in the art.
  • the term generally refers a nucleotide sequence that is shared by one or more polynucleotide sequences, such as genes, gene transcripts and/or non-coding polynucleotides.
  • a homologous sequence can be a nucleotide sequence that is shared by two or more genes encoding related but different proteins, such as different members of a gene family, different protein epitopes, different protein isoforms or completely divergent genes.
  • a homologous sequence can be a nucleotide sequence that is shared by two or more non- coding polynucleotides, such as noncoding DNA or RNA, regulatory sequences, introns, and sites of transcriptional control or regulation. Homologous sequences can also include sequence regions shared by more than one polynucleotide sequence. Homology does not need to be perfect identity (100%), as partially homologous sequences are also contemplated by and within the scope of the instant invention ⁇ e.g., at least 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% etc.). Percent homology is the number of matching nucleotides between two sequences divided by the total length being compared, multiplied by 100.
  • RNAi activity refers to an increase in RNAi activity measured in vitro and/or in vivo, where the RNAi activity is a reflection of both the ability of the siNA to mediate RNAi and the stability of the siNAs of the invention.
  • the product of these activities can be increased in vitro and/or in vivo compared to an all RNA siNA or an siNA containing a plurality of ribonucleotides.
  • the activity or stability of the siNA molecule can be decreased ⁇ i.e., less than ten-fold), but the overall activity of the siNA molecule is enhanced in vitro and/or in vivo.
  • inhibitor refers to its meaning as is generally accepted in the art.
  • RNA molecules generally refers the reduction in the expression of the gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or SIR-ONC-00001 activity of one or more proteins or protein subunits, below that observed in the absence of the nucleic acid molecules (e.g. , siNA) of the invention.
  • Down-regulation can also be associated with post-transcriptional silencing, such as, RNAi mediated cleavage or by alteration in DNA methylation patterns or DNA chromatin structure.
  • Inhibition, down-regulation or reduction with an siNA molecule can be in reference to an inactive molecule, an attenuated molecule, an siNA molecule with a scrambled sequence, or an siNA molecule with mismatches or alternatively, it can be in reference to the system in the absence of the nucleic acid.
  • inhibitors of cell proliferation and survival signaling pathway refers to pharmaceutical agents that inhibit cell surface receptors and signal transduction cascades downstream of those surface receptors.
  • integrin blockers refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the ⁇ ⁇ ⁇ 3 integrin, to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the ⁇ ⁇ 5 integrin, to compounds which antagonize, inhibit or counteract binding of a physiological ligand to both the ⁇ ⁇ ⁇ 3 integrin and the ⁇ ⁇ 5 integrin, and to compounds which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells.
  • the term also refers to antagonists of the ⁇ 2 ⁇ ⁇ 5 ⁇ ⁇ 6 ⁇ and ⁇ 6 ⁇ 4 integrins.
  • the term also refers to antagonists of any combination of ⁇ ⁇ ⁇ 3 , ⁇ ⁇ 5, ⁇ 2 ⁇ ⁇ 5 ⁇ ⁇ 6 ⁇ and ⁇ 6 ⁇ 4 integrins.
  • internucleoside linkage or “internucleoside linker” or “internucleotide linkage” or “internucleotide linker” are used herein interchangeably and refer to any linker or linkage between two nucleoside units, as is known in the art, including, for example, but not limitation, phosphate, analogs of phosphate, phosphonate, guanidium, hydroxylamine, hydroxylhydrazinyl, amide, carbamate, alkyl, and substituted alkyl linkages.
  • the internucleoside linkages constitute the backbone of a nucleic acid molecule.
  • mammalian or “mammal” as used herein refers to its meaning as is generally accepted in the art.
  • the term generally refers to any warm blooded vertebrate species, such as a human, mouse, rat, dog, cat, hamster, guinea pig, rabbit, livestock, and the like.
  • MDI tered dose inhaler
  • MDI systems includes a suitable channeling device. Suitable channeling devices comprise for example, a valve actuator and a cylindrical or cone-like passage through which medicament can be delivered from the filled canister via the metering valve to the nose or mouth of a patient such as a mouthpiece actuator.
  • microRNA or "miRNA” as used herein refers to its meaning as is generally accepted in the art.
  • the term generally refers a small double- stranded RNA that regulates the expression of target messenger RNAs either by mRNA cleavage, translational repression/inhibition or heterochromatic silencing (see for example Ambros, 2004, Nature, 431, 350-355; Bartel, 2004, Cell, 116, 281-297; Cullen, 2004, Virus Research., 102, 3-9; He et al, 2004, Nat. Rev. Genet., 5, 522-531; Ying et al, 2004, Gene, 342, 25-28; and Sethupathy et al, 2006, RNA, 12: 192-197).
  • modulate refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to when the expression of a gene, or level of one or more RNA molecules (coding or non-coding), or activity of one or more RNA molecules or proteins or protein subunits, is up-regulated or down-regulated, such that expression, level, or activity is greater than or less than that observed in the absence of the molecule that effects modulation.
  • the term “modulate” in some embodiments can refer to inhibition and in other embodiments can refer to potentiation or up-regulation, e.g., of gene expression.
  • modified nucleotide refers to its meaning as is generally accepted in the art.
  • the term generally refers a nucleotide, which contains a modification in the chemical structure of the base, sugar and/or phosphate of the unmodified (or natural) nucleotide as is generally known in the art.
  • modified nucleotides are described herein and in U.S. Application No. 12/064,014. SIR-ONC-00001
  • NSAIDs that are selective COX-2 inhibitors refers to NSAIDs, which possess a specificity for inhibiting COX-2 over COX-1 of at least 100 fold as measured by the ratio of IC50 for COX-2 over IC50 for COX-1 evaluated by cell or microsomal assays.
  • non-base paired refers to nucleotides that are not base paired between the sense strand or sense region and the antisense strand or antisense region of an double- stranded siNA molecule; and can include for example, but not limitation, mismatches, overhangs, single stranded loops, etc.
  • non-nucleotide refers to any group or compound which can be incorporated into a nucleic acid chain in the place of one or more nucleotide units, such as for example but not limitation abasic moieties or alkyl chains.
  • the group or compound is "abasic" in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine and therefore lacks a nucleobase at the 1 '-position.
  • nucleotide is used as is generally recognized in the art. Nucleotides generally comprise a nucleobase, a sugar, and an internucleoside linkage, e.g., a phosphate.
  • the base can be a natural bases (standard), modified bases, or a base analog, as are well known in the art. Such bases are generally located at the ⁇ position of a nucleotide sugar moiety.
  • nucleotides can be unmodified or modified at the sugar, internucleoside linkage, and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and others; see, for example, U.S. Application No. 12/064,014.
  • overhang refers to its meaning as is generally accepted in the art. With reference to exemplary double stranded nucleic acid molecules, the term generally refers to the terminal portion of a nucleotide sequence that is not base paired between the two strands of a double- stranded nucleic acid molecule (see for example, Figure 4). Overhangs, when present, are typically at the 3'-end of one or both strands in a siNA duplex.
  • parenteral refers to its meaning as is generally accepted in the art.
  • the term generally refers methods or techniques of administering a molecule, drug, SIR-ONC-00001 agent, or compound in a manner other than through the digestive tract, and includes epicutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, or intrathecal injection or infusion techniques and the like.
  • pathway target refers to any target involved in pathways of gene expression or activity.
  • any given target can have related pathway targets that can include upstream, downstream, or modifier genes in a biologic pathway.
  • pathway target genes can provide additive or synergistic effects in the treatment of diseases, conditions, and traits herein.
  • phosphorothioate refers to an intemucleotide phosphate linkage comprising one or more sulfur atoms in place of an oxygen atom.
  • phosphorothioate refers to both phosphorothioate and phosphorodithioate intemucleotide linkages.
  • Prenyl-protein transferase inhibitor refers to a compound that inhibits any one or any combination of the prenyl-protein transferase enzymes, including farnesyl-protein transferase (FPTase), geranylgeranyl-protein transferase type I (GGPTase-I), and geranylgeranyl-protein transferase type-II (GGPTase-II, also called Rab GGPTase).
  • FPTase farnesyl-protein transferase
  • GGPTase-I geranylgeranyl-protein transferase type I
  • GGPTase-II geranylgeranyl-protein transferase type-II
  • retinoid receptor modulators refers to compounds that interfere or inhibit the binding of retinoids to the receptor, regardless of mechanism.
  • ribonucleotide refers to its meaning as is generally accepted in the art.
  • the term generally refers to a nucleotide with a hydroxyl group at the 2' position of a ⁇ -D-ribofuranose moiety.
  • RNA refers to its generally accepted meaning in the art. Generally, the term RNA refers to a molecule comprising at least one ribofuranoside moiety. The term can include double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides.
  • Such alterations can include addition of non-nucleotide material, such as to the end(s) of the siNA or internally, for example at one or SIR-ONC-00001 more nucleotides of the RNA.
  • Nucleotides in the RNA molecules of the instant invention can also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.
  • RNA interference refers to the biological process of inhibiting or down regulating gene expression in a cell, as is generally known in the art, and which is mediated by short interfering nucleic acid molecules, see for example Zamore and Haley, 2005, Science, 309, 1519-1524; Vaughn and Martienssen, 2005, Science, 309, 1525-1526; Zamore et al, 2000, Cell, 101, 25-33; Bass, 2001, Nature, 411, 428-429; Elbashir et al, 2001, Nature, 411, 494-498; and Kreutzer et al, International PCT Publication No.
  • RNAi is meant to be equivalent to other terms used to describe sequence specific RNA interference, such as post transcriptional gene silencing, translational inhibition, transcriptional inhibition, or epigenetics.
  • siNA molecules of the invention can be used to epigenetically silence genes at either the post- transcriptional level or the pre-transcriptional level.
  • epigenetic modulation of gene expression by siNA molecules of the invention can result from siNA mediated modification of chromatin structure or methylation patterns to alter gene expression (see, for example, Verdel et al, 2004, Science, 303, 672-676; Pal-Bhadra et al, 2004, Science, 303, 669-672; Allshire, 2002, Science, 297, 1818-1819; Volpe et al, 2002, Science, 297, 1833- 1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al, 2002, Science, 297, 2232-2237).
  • modulation of gene expression by siNA molecules of the invention can result from siNA mediated cleavage of RNA (either coding or non-coding RNA) via RISC, or via translational inhibition, as is known in the art or modulation can result from SIR-ONC-00001 transcriptional inhibition (see for example Janowski et al, 2005, Nature Chemical Biology, 1, 216-222).
  • RNAi inhibitor refers to any molecule that can down regulate, reduce or inhibit RNA interference function or activity in a cell or organism.
  • An RNAi inhibitor can down regulate, reduce or inhibit RNAi (e.g., RNAi mediated cleavage of a target polynucleotide, translational inhibition, or transcriptional silencing) by interaction with or interfering with the function of any component of the RNAi pathway, including protein components such as RISC, or nucleic acid components such as miRNAs or siRNAs.
  • a RNAi inhibitor can be an siNA molecule, an antisense molecule, an aptamer, or a small molecule that interacts with or interferes with the function of RISC, a miRNA, or an siRNA or any other component of the RNAi pathway in a cell or organism.
  • RNAi e.g., RNAi mediated cleavage of a target polynucleotide, translational inhibition, or transcriptional silencing
  • a RNAi inhibitor of the invention can be used to modulate (e.g., up-regulate or down regulate) the expression of a target gene.
  • sense region refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a nucleotide sequence of an siNA molecule having complementarity to an antisense region of the siNA molecule.
  • the sense region of an siNA molecule can comprise a nucleic acid sequence having homology or sequence identity with a target nucleic acid sequence. In one embodiment, the sense region of the siNA molecule is also referred to as the sense strand or passenger strand.
  • short interfering nucleic acid refers to any nucleic acid molecule capable of inhibiting or down regulating gene expression or viral replication by mediating RNA interference ("RNAi") or gene silencing in a sequence- specific manner. These terms can refer to both individual nucleic acid molecules, a plurality of such nucleic acid molecules, or pools of such nucleic acid molecules.
  • RNAi RNA interference
  • the siNA can be a double-stranded nucleic acid molecule comprising self-complementary sense and antisense strands, wherein the antisense SIR-ONC-00001 strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense strand comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof.
  • the siNA can be a polynucleotide with a duplex, asymmetric duplex, hairpin or asymmetric hairpin secondary structure, having self-complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence in a separate target nucleic acid molecule or a portion thereof and the sense region comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof.
  • the siNA can be a circular single-stranded polynucleotide having two or more loop structures and a stem comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof, and wherein the circular polynucleotide can be processed either in vivo or in vitro to generate an active siNA molecule capable of mediating RNAi.
  • the siNA can also comprise a single-stranded polynucleotide having a nucleotide sequence complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof (for example, where such siNA molecule does not require the presence within the siNA molecule of a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof), wherein the single-stranded polynucleotide can further comprise a terminal phosphate group, such as a 5'-phosphate (see for example, Martinez et ah, 2002, Cell, 110, 563-574 and Schwarz et al., 2002, Molecular Cell, 10, 537-568), or 5',3'- diphosphate.
  • a terminal phosphate group such as a 5'-phosphate (see for example, Martinez et ah, 2002, Cell, 110, 563-574 and Schwarz et al., 2002, Molecular Cell, 10, 537-568), or 5',3'- di
  • subject refers to its meaning as is generally accepted in the art.
  • the term generally refers an organism to which the nucleic acid molecules of the invention can be administered.
  • a subject can be a mammal or mammalian cells, including a human or human cells.
  • the term also refers to an organism, which is a donor or recipient of explanted cells or the cells themselves.
  • systemic administration refers to its meaning as is generally accepted in the art.
  • the term generally refers in vivo systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body. SIR-ONC-00001
  • target refers to any MET target protein, peptide, or polypeptide, such as encoded by Genbank Accession Nos. shown in Table 10.
  • the term also refers to nucleic acid sequences or target polynucleotide sequence encoding any target protein, peptide, or polypeptide, such as proteins, peptides, or polypeptides encoded by sequences having Genbank Accession Nos. shown in Table 10.
  • the target of interest can include target polynucleotide sequences, such as target DNA or target RNA.
  • target is also meant to include other sequences, such as differing isoforms, mutant target genes, splice variants of target polynucleotides, target polymorphisms, and non-coding (e.g., ncRNA, miRNA, stRNA, sRNA) or other regulatory polynucleotide sequences as described herein.
  • non-coding e.g., ncRNA, miRNA, stRNA, sRNA
  • target site refers to its meaning as is generally accepted in the art.
  • the term generally refers to a sequence within a target nucleic acid molecule, (e.g., RNA) that is “targeted”, e.g., for cleavage mediated by an siNA construct, which contains sequences within its antisense region that are complementary to the target sequence.
  • terapéuticaally effective amount refers to its meaning as is generally accepted in the art.
  • the term generally refers to the amount of the compound or composition that will elicit the biological or medical response of a cell, tissue, system, animal or human that is be sought by the researcher, veterinarian, medical doctor or other clinician. For example, if a given clinical treatment is considered effective when there is at least a 25% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is that amount necessary to effect at least a 25% reduction in that parameter.
  • universal base refers to its meaning as is generally accepted in the art.
  • the term universal base generally refers to nucleotide base analogs that form base pairs with each of the natural DNA/RNA bases with little or no discrimination between them.
  • Non-limiting examples of universal bases include C-phenyl, C-naphthyl and other aromatic derivatives, inosine, azole carboxamides, and nitroazole derivatives such as 3- nitropyrrole, 4-nitroindole, 5-nitroindole, and 6-nitroindole as known in the art (see for example, Loakes, 2001, Nucleic Acids Research, 29, 2437-2447). SIR-ONC-00001
  • up-regulate refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to an increase in the expression of a gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or activity of one or more RNAs, proteins or protein subunits, above that observed in the absence of the nucleic acid molecules (e.g. , siNA) of the invention. In certain instances, up-regulation or promotion of gene expression with an siNA molecule is above that level observed in the presence of an inactive or attenuated molecule.
  • siNA nucleic acid molecules
  • up-regulation or promotion of gene expression with siNA molecules is above that level observed in the presence of, for example, an siNA molecule with scrambled sequence or with mismatches.
  • up-regulation or promotion of gene expression with a nucleic acid molecule of the instant invention is greater in the presence of the nucleic acid molecule than in its absence.
  • up-regulation or promotion of gene expression is associated with inhibition of RNA mediated gene silencing, such as RNAi mediated cleavage or silencing of a coding or non-coding RNA target that down regulates, inhibits, or silences the expression of the gene of interest to be up-regulated.
  • the down regulation of gene expression can, for example, be induced by a coding RNA or its encoded protein, such as through negative feedback or antagonistic effects.
  • the down regulation of gene expression can, for example, be induced by a non-coding RNA having regulatory control over a gene of interest, for example by silencing expression of the gene via translational inhibition, chromatin structure, methylation, RISC mediated RNA cleavage, or translational inhibition.
  • inhibition or down regulation of targets that down regulate, suppress, or silence a gene of interest can be used to up- regulate expression of the gene of interest toward therapeutic use.
  • vector refers to its meaning as is generally accepted in the art.
  • vector generally refers to any nucleic acid- and/or viral-based expression system or technique used to deliver one or more nucleic acid molecules.
  • the present invention provides compositions and methods comprising siNAs targeted to MET that can be used to treat diseases, e.g., malignancies and/or cancers associated with MET expression.
  • the nucleic acid molecules SIR-ONC-00001 of the invention comprise at least a 15 nucleotide sequence of the sequences shown in Table la and Table lb.
  • the siNAs can be provided in several forms.
  • the siNA can be isolated as one or more siNA compounds, or it may be in the form of a transcriptional cassette in a DNA plasmid.
  • the siNA may also be chemically synthesized and can include modifications as shown, for example, but not limitation, in Table lc and Table 11.
  • the siNAs can be administered alone or co-administered with other siNA molecules or with conventional agents that treat a MET related disease or condition.
  • the siNA molecules of the invention can be used to mediate gene silencing, specifically MET, via interaction with RNA transcripts or alternately by interaction with particular gene sequences, wherein such interaction results in modulation of gene silencing either at the transcriptional level or post-transcriptional level such as, for example, but not limited to, RNAi or through cellular processes that modulate the chromatin structure or methylation patterns of the target and prevent transcription of the target gene, with the nucleotide sequence of the target thereby mediating silencing.
  • the target is any of MET RNA, DNA, or mRNA
  • the invention provides short interfering nucleic acid (siNA) molecules for inhibiting the expression of the MET gene in a cell or mammal.
  • the siNA can be single- stranded or double- stranded. When double- stranded, the siNA comprising a sense and an antisense stand.
  • the antisense strand is complementary to at least a part of an mRNA formed in the expression of the MET gene.
  • the sense strand comprises a region that is complementary to the antisense strand.
  • the antisense strand comprises at least a 15 nucleotide sequence of an antisense sequence listed in Table lb.
  • the double- stranded siNA comprises at least a 15 nucleotide sequence of the sense strand in Table lb and at least a 15 nucleotide sequence of the antisense strand in Table lb.
  • One or more of the nucleotides of the siNAs of the invention are optionally modified.
  • some siNAs of the invention comprises at least one nucleotide sequence selected from the groups of sequences provide in Table lc.
  • the siNA comprises at least two sequences selected from the group of sequences provided in Table lc, wherein one of the at least two sequences is complementary to another of the at least two sequences and one of the at least two sequences is complementary to a sequence of a mRNA generated in the SIR-ONC-00001 expression of the MET gene.
  • Examples of certain modified siNAs of the invention are in Table lc.
  • the double stranded RNA molecules of the invention can comprise two distinct and separate strands that can be symmetric or asymmetric and are complementary, i.e., two single- stranded RNA molecules, or can comprise one single- stranded molecule in which two complementary portions, e.g., a sense region and an antisense region, are base-paired, and are covalently linked by one or more single- stranded "hairpin" areas ⁇ i.e. loops) resulting in, for example, a single-stranded short-hairpin polynucleotide or a circular single-stranded polynucleotide.
  • the linker can be polynucleotide linker or a non-nucleotide linker. In some embodiments, the linker is a non-nucleotide linker.
  • a hairpin or circular siNA molecule of the invention contains one or more loop motifs, wherein at least one of the loop portions of the siNA molecule is biodegradable. For example, a single-stranded hairpin siNA molecule of the invention is designed such that degradation of the loop portion of the siNA molecule in vivo can generate a double- stranded siNA molecule with 3'-terminal overhangs, such as 3'-terminal nucleotide overhangs comprising 1, 2, 3 or 4 nucleotides.
  • a circular siNA molecule of the invention is designed such that degradation of the loop portions of the siNA molecule in vivo can generate a double-stranded siNA molecule with 3'-terminal overhangs, such as 3 '-terminal nucleotide overhangs comprising about 2 nucleotides.
  • each strand, the sense (passenger) strand and antisense (guide) strand are independently about 15 to about 30 ⁇ e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length.
  • each strand of the symmetric siNA molecules of the invention are about 19-24 ⁇ e.g., about 19, 20, 21, 22, 23 or 24) nucleotides in length.
  • the antisense region or strand of the molecule is about 15 to about 30 ⁇ e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length, wherein the sense region is about 3 to about 25 ⁇ e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides in length.
  • each strand of the asymmetric siNA molecules of the invention is about 19-24 (e.g., about 19, 20, 21, 22, 23 or 24) nucleotides in length.
  • siNA molecules of the invention comprise single stranded hairpin siNA molecules, wherein the siNA molecules are about 25 to about 70 (e.g. , about 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 40, 45, 50, 55, 60, 65, or 70) nucleotides in length.
  • siNA molecules of the invention comprise single-stranded circular siNA molecules, wherein the siNA molecules are about 38 to about 70 (e.g., about 38, 40, 45, 50, 55, 60, 65, or 70) nucleotides in length.
  • siNA molecules of the invention comprise single-stranded non-circular siNA molecules, wherein the siNA molecules are independently about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length.
  • the siNA duplexes of the invention independently comprise about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) base pairs.
  • the duplex structure of siNAs of the invention is between 15 and 30, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 base pairs in length.
  • the siNA molecules of the invention comprise about 3 to 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) base pairs.
  • the duplex structure of siNAs of the invention is between 15 and 25, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 base pairs in length.
  • the siNA molecules of the invention are hairpin or circular structures
  • the siNA molecules comprise about 15 to about 30 (e.g. , about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) base pairs.
  • the sense strand and antisense strand, or the sense region and antisense region, of the siNA molecules of the invention can be complementary.
  • the antisense strand or antisense SIR-ONC-00001 region can be complementary to a nucleotide sequence or a portion thereof of the MET target RNA.
  • the sense strand or sense region of the siNA can comprise a nucleotide sequence of a MET gene or a portion thereof.
  • the sense region or sense strand of an siNA molecule of the invention is complementary to that portion of the antisense region or antisense strand of the siNA molecule that is complementary to a MET target polynucleotide sequence, such as for example, but not limited to, those sequences represented by GENBANK Accession Nos. shown in Table 10.
  • siNA molecules of the invention have perfect complementarity between the sense strand or sense region and the antisense strand or antisense region of the siNA molecule.
  • the antisense strand of the siNA molecules of the invention are perfectly complementary to a corresponding target nucleic acid molecule.
  • siNA molecules of the invention have partial complementarity (i.e., less than 100% complementarity) between the sense strand or sense region and the antisense strand or antisense region of the siNA molecule or between the antisense strand or antisense region of the siNA molecule and a corresponding target nucleic acid molecule.
  • the double- stranded nucleic acid molecules of the invention have between about 15 to about 30 (e.g. , about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in one strand that are complementary to the nucleotides of the other strand.
  • the molecules have between about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in the sense region that are complementary to the nucleotides of the antisense region, of the double-stranded nucleic acid molecule.
  • the double- stranded nucleic acid molecules of the invention have between about 15 to about 30 (e.g. , about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in the antisense strand that are complementary to a nucleotide sequence of its corresponding target nucleic acid molecule.
  • the siNA molecule can contain one or more nucleotide deletions, substitutions, mismatches and/or additions; provided, however, that the siNA molecule maintains its activity, for example, to mediate RNAi.
  • the deletion, SIR-ONC-00001 substitution, mismatch and/or addition can result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair.
  • the double-stranded nucleic acid molecules of the invention have 1 or more (e.g., 1, 2, 3, 4, 5, or 6) nucleotides, in one strand or region that are mismatches or non-base-paired with the other strand or region.
  • the double- stranded nucleic acid molecules of the invention have 1 or more (e.g., 1, 2, 3, 4, 5, or 6) nucleotides in each strand or region that are mismatches or non-base-paired with the other strand or region.
  • the siNA of the invention contains no more than 3 mismatches. If the antisense strand of the siNA contains mismatches to a target sequence, it is preferable that the area of mismatch not be located in the center of the region of complementarity.
  • the siNA molecule can contain one or more nucleotide deletions, substitutions, mismatches and/or additions to a sequence in Table lb provided, however, that the siNA molecule maintains its activity, for example, to mediate RNAi.
  • the deletion, substitution, mismatch and/or addition can result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair.
  • the invention also comprises double- stranded nucleic acid (siNA) molecules as otherwise described hereinabove in which the first strand and second strand are complementary to each other and wherein at least one strand is hybridisable to the polynucleotide sequence of a sequence in Table lb under conditions of high stringency, and wherein any of the nucleotides is unmodified or chemically modified.
  • siNA double- stranded nucleic acid
  • Hybridization techniques are well known to the skilled artisan (see for instance, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)).
  • Preferred stringent hybridization conditions include overnight incubation at 42°C in a solution comprising: 50% formamide, 5xSSC (150mM NaCl, 15mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution, 10% dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters in O. lx SSC at about 65°C.
  • the first strand has about 15, 16, 17, 18, 19, 20 or 21 nucleotides that are complementary to the nucleotides of the other strand and at least one strand SIR-ONC-00001 is hybridisable to a polynucleotide sequence in Table lb.
  • the first strand has about 15, 16, 17, 18, 19, 20 or 21 nucleotides that are complementary to the nucleotides of the other strand and at least one strand is hybridisable to SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; under conditions of high stringency, and wherein any of the nucleotides is unmodified or chemically modified.
  • the siNA molecules of the invention comprise overhangs of about 1 to about 4 (e.g. , about 1, 2, 3 or 4) nucleotides.
  • the nucleotides in the overhangs can be the same or different nucleotides.
  • the overhangs occur at the 3 '-end at one or both strands of the double-stranded nucleic acid molecule.
  • a double- stranded nucleic acid molecule of the invention can comprise a nucleotide or non-nucleotide overhang at the 3 '-end of the antisense strand/region, the 3 '-end of the sense strand/region, or both the antisense strand/region and the sense strand/region of the double-stranded nucleic acid molecule.
  • the nucleotides comprising the overhang portion of an siNA molecule of the invention comprise sequences based on the MET target polynucleotide sequence in which nucleotides comprising the overhang portion of the antisense strand/region of an siNA molecule of the invention can be complementary to nucleotides in the MET target polynucleotide sequence and/or nucleotides comprising the overhang portion of the sense strand/region of an siNA molecule of the invention can comprise the nucleotides in the MET target polynucleotide sequence.
  • the overhang comprises a two nucleotide overhang that is complementary to a portion of the MET target polynucleotide sequence. In other embodiments, however, the overhang comprises a two nucleotide overhang that is not complementary to a portion of the MET target polynucleotide sequence. In certain embodiments, the overhang comprises a 3'-UU overhang that is not complementary to a portion of the MET target polynucleotide sequence. In other embodiments, the overhang comprises a UU overhang at the 3' end of the antisense strand and a TT overhang at the 3' end of the sense strand. In other embodiments, the overhang comprises nucleotides as described in the examples, Tables, and Figures herein. SIR-ONC-00001
  • the overhangs are optionally chemically modified at one or more nucleic acid sugar, base, or backbone positions.
  • modified nucleotides in the overhang portion of a double- stranded nucleic acid (siNA) molecule of the invention include: 2'-0-alkyl (e.g., 2'-0-methyl), 2'-deoxy, 2'-deoxy-2'-fluoro, 2'-deoxy-2'- fluoroarabino (FANA), 4'-thio, 2'-0-trifluoromethyl, 2'-0-ethyl-trifluoromethoxy, 2'-0- difluoromethoxy-ethoxy, universal base, acyclic, or 5-C-methyl nucleotides.
  • the overhang nucleotides are each independently, a 2'-0-alkyl nucleotide, a 2'-0- methyl nucleotide, a 2'-dexoy-2-fluoro nucleotide, or a 2'-deoxy ribonucleotide. In some instances the overhang nucleotides are linked by a one or more phosphorothioate linkages.
  • siNA molecules of the invention comprise duplex nucleic acid molecules with blunt ends (i.e., without nucleotide overhangs), where both ends are blunt, or alternatively, where one of the ends is blunt.
  • the siNA molecules of the invention can comprises one blunt end, for example wherein the 5 '-end of the antisense strand and the 3 '-end of the sense strand do not have any overhanging nucleotides.
  • the siNA molecule comprises one blunt end, for example wherein the 3 '-end of the antisense strand and the 5 '-end of the sense strand do not have any overhanging nucleotides.
  • siNA molecules of the invention comprise two blunt ends, for example wherein the 3 '-end of the antisense strand and the 5 '-end of the sense strand as well as the 5 '-end of the antisense strand and 3 '-end of the sense strand do not have any overhanging nucleotides.
  • the sense strand and/or the antisense strand can further have a cap, such as described herein or as known in the art, at the 3'-end, the 5'-end, or both of the 3' and 5'-ends of the sense strand and/or antisense strand.
  • the cap can be at either one or both of the terminal nucleotides of the polynucleotide.
  • the cap is at one of both of the ends of the sense strand of a double- stranded siNA molecule.
  • the cap is at the 3 '-end of antisense (guide) strand. In preferred embodiments, the caps are at the 3'-end of the sense strand and the 5'-end of the sense strand.
  • terminal caps include an inverted abasic nucleotide, an inverted deoxy abasic nucleotide, an inverted nucleotide moiety, a group shown in Figure 5, a glyceryl modification, an alkyl or cycloalkyl group, a heterocycle, or any other cap as is generally known in the art.
  • any of the embodiments of the siNA molecules of the invention can have a 5' phosphate termini.
  • the siNA molecules lack terminal phosphates.
  • siNA molecule or construct of the invention can comprise one or more chemical modifications. Modifications can be used to improve in vitro or in vivo characteristics such as stability, activity, toxicity, immune response (e.g., prevent stimulation of an interferon response, an inflammatory or pro -inflammatory cytokine response, or a Toll-like Receptor (TIF) response), and/or bioavailability.
  • TNF Toll-like Receptor
  • RNAi activity RNAi activity
  • stability RNAi activity
  • Various chemically modified siNA motifs disclosed herein provide the capacity to maintain RNAi activity that is substantially similar to unmodified or minimally modified active siRNA (see for example Elbashir et al., 2001, EMBO J., 20:6877-6888) while at the same time providing nuclease resistance and pharmacokinetic properties suitable for use in therapeutic applications.
  • the siNA molecules of the invention comprise modifications wherein any (e.g., one or more or all) nucleotides present in the sense and/or antisense strand are modified nucleotides (e.g., wherein one nucleotide is modified, some nucleotides (i.e., plurality or more than one) are modified, or all nucleotides are modified nucleotides.
  • the siNA molecules of the invention are partially modified (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 55, or 59 nucleotides are modified) with chemical modifications.
  • an siNA molecule of the invention comprises at least about 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, or 60 nucleotides that are modified nucleotides.
  • the siNA molecules of the invention are completely modified (e.g., 100% modified) with chemical modifications, i.e., the siNA molecule does not contain any SIR-ONC-00001 ribonucleotides.
  • one or more of the nucleotides in the sense strand of the siNA molecules of the invention are modified.
  • one or more of the nucleotides in the antisense strand of the siNA molecules of the invention are modified.
  • the chemical modification within a single siNA molecule can be the same or different.
  • at least one strand has at least one chemical modification.
  • each strand has at least one chemical modifications, which can be the same or different, such as, sugar, base, or backbone (i.e., internucleotide linkage) modifications.
  • siNA molecules of the invention contain at least 2, 3, 4, 5, or more different chemical modifications.
  • Non-limiting examples of chemical modifications that are suitable for use in the present invention are disclosed in U.S. Patent Applications Nos. 10/444,853; 10/981,966; 12/064,014 and in references cited therein and include sugar, base, and phosphate, non- nucleotide modifications, and/or any combination thereof.
  • At least one modified nucleotide is a 2'-deoxy-2-fluoro nucleotide, a 2'-deoxy nucleotide, a 2'-0-alkyl (e.g., 2'-0-methyl) nucleotide, or a locked nucleic acid (LNA) nucleotide as is generally recognized in the art.
  • LNA locked nucleic acid
  • At least one nucleotide has a ribo-like, Northern or A form helix configuration (see e.g., Saenger, Principles of Nucleic Acid Structure, Springer- Verlag ed., 1984).
  • Non-limiting examples of nucleotides having a Northern configuration include locked nucleic acid (LNA) nucleotides (e.g., 2'-0, 4'-C-methylene-(D- ribofuranosyl) nucleotides); 2'-methoxyethoxy (MOE) nucleotides; 2'-methyl-thio-ethyl nucleotides, 2'-deoxy-2'-fluoro nucleotides; 2'-deoxy-2'-chloro nucleotides; 2'-azido nucleotides; 2'-0-trifluoromethyl nucleotides; 2'-0-ethyl-trifluoromethoxy nucleotides; 2'-0- difluoromethoxy-ethoxy nucleotides; 4'-thio nucleotides and 2'-0-methyl nucleotides.
  • LNA locked nucleic acid
  • MOE 2'-methoxyethoxy
  • a majority (e.g., greater than 50%) of the pyrimidine nucleotides present in the double- stranded siNA molecule comprises a sugar modification.
  • a majority (e.g., greater than 50%) of the purine nucleotides present in the double-stranded siNA molecule comprises a sugar modification.
  • the pyrimidine nucleotides in the antisense strand are 2'-0- methyl or 2'-deoxy-2'-fluoro pyrimidine nucleotides and the purine nucleotides present in the antisense strand are 2'-0-methyl nucleotides or 2'-deoxy nucleotides.
  • the pyrimidine nucleotides in the sense strand are 2'-deoxy-2'-fluoro pyrimidine nucleotides and the purine nucleotides present in the sense strand are 2'-0-methyl or 2'-deoxy purine nucleotides.
  • all the pyrimidine nucleotides in the complementary region on the sense strand are 2'-deoxy-2'-fluoro pyrimidine nucleotides. In certain embodiments, all of the pyrimidine nucleotides in the complementary region of the antisense strand are 2'-deoxy-2'-fluoro pyrimidine nucleotides. In certain embodiments, all the purine nucleotides in the complementary region on the sense strand are 2'-deoxy purine nucleotides. In certain embodiments, all of the purines in the complementary region on the antisense strand are 2'-0-methyl purine nucleotides.
  • all of the pyrimidine nucleotides in the complementary regions on the sense strand are 2'-deoxy-2'-fluoro pyrimidine nucleotides; all of the pyrimidine nucleotides in the complementary region of the antisense strand are 2'-deoxy-2'-fluoro pyrimidine nucleotides; all the purine nucleotides in the complementary region on the sense strand are 2'-deoxy purine nucleotides and all of the purines in the complementary region on the antisense strand are 2'-0-methyl purine nucleotides.
  • At least 5 or more of the pyrimidine nucleotides in one or both stands are 2' -deoxy-2' -fluoro pyrimidine nucleotides. In some embodiments, at least 5 or more of the pyrimidine nucleotides in one or both stands are 2'-0-methyl pyrimidine nucleotides. In some embodiments, at least 5 or more of the purine nucleotides in one or both stands are 2'- deoxy-2' -fluoro purine nucleotides In some embodiments, at least 5 or more of the purine nucleotides in one or both stands are 2'-0-methyl purine nucleotides.
  • the purines and pyrimidines are differentially modified at the 2'-sugar position (i.e., at least one purine has a different modification from at least one pyrimidine in the same or different strand at the 2'-sugar position).
  • at least 5 or more of the pyrimidine nucleotides in one or both stands are 2' -deoxy-2' - fluoro pyrimidine nucleotides and at least 5 or more purine nucleotides in one or both strands are 2'-0-methyl purine nucleotides.
  • At least 5 or more of the pyrimidine SIR-ONC-00001 nucleotides in one or both stands are 2'-0-methyl pyrimidine nucleotides and at least 5 or more purine nucleotides in one or both strands are 2'-deoxy-2'-fluoro purine nucleotides.
  • the modified siNA molecules of the invention can comprise modifications at various locations within the siNA molecule.
  • the double-stranded siNA molecule of the invention comprises modified nucleotides at internal base paired positions within the siNA duplex.
  • a double- stranded siNA molecule of the invention comprises modified nucleotides at non-base paired or overhang regions of the siNA molecule.
  • a double- stranded siNA molecule of the invention comprises modified nucleotides at terminal positions of the siNA molecule. For example, such terminal regions include the 3'- position and/or 5 '-position of the sense and/or antisense strand or region of the siNA molecule.
  • any of the modified siNA molecules of the invention can have a modification in one or both oligonucleotide strands of the siNA duplex, for example in the sense strand, the antisense strand, or both strands.
  • each strand of the double-stranded siNA molecules of the invention can have one or more chemical modifications, such that each strand comprises a different pattern of chemical modifications.
  • each strand of a double-stranded siNA molecule of the invention comprises a different pattern of chemical modifications, such as any Stab modification chemistries described herein (see Table 11) or any combination thereof, i.e., different combinations of defined Stabilzation chemistry (Stab) sense and antisense strands.
  • Stab Stabilzation chemistry
  • the stabilization chemistries referred to in Table 11 as Stab can be combined in any combination of sense/antisense chemistries, such as Stab 7/8, Stab 7/11, Stab 8/8, Stab 18/8, Stab 18/11, Stab 12/13, Stab 7/13, Stab 18/13, Stab 7/19, Stab 8/19, SIR-ONC-00001
  • one or more (for example 1, 2, 3, 4 or 5) nucleotides at the 5 '-end of the guide strand or guide region (also known as antisense strand or antisense region) of the siNA molecule are ribonucleotides.
  • the present invention provides a double- stranded short interfering nucleic acid (siNA) molecule that modulates the expression of MET, wherein the siNA comprises a sense strand and an antisense strand; each strand is independently 15 to 30 nucleotides in length; and the antisense strand comprises at least 15 nucleotides having sequence complementary to any of:
  • the antisense strand of a siNA molecule of the invention comprises at least a 15 nucleotide sequence of:
  • the sense strand of a siNA molecule of the invention comprises at least a 15 nucleotide sequence of:
  • a siNA molecule of the invention comprises any of:
  • the nucleotides of the at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101 form a contiguous stretch of nucleotides.
  • the siNA molecule can contain one or more nucleotide deletions, substitutions, mismatches and/or additions to the at least 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; provided, however, that the siNA molecule maintains its activity, for example, to mediate RNAi.
  • the deletion, substitution, mismatch and/or addition can result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair.
  • double- stranded siNA molecules are provided, wherein the molecule has a sense strand and an antisense strand and comprises the following formula (A): SIR-ONC-00001
  • the upper strand is the sense strand and the lower strand is the antisense strand of the double- stranded nucleic acid molecule; wherein the antisense strand comprises at least a 15 nucleotide sequence of SEQ ID NO: 1049, SEQ ID NO: 1091, SEQ ID NO: 1099, or SEQ ID NO: 1101, and the sense strand comprises a sequence having complementarity to the antisense strand; each N is independently a nucleotide which is unmodified or chemically modified or a non-nucleotide; each B is a terminal cap that is present or absent;
  • (N) represents overhanging nucleotides, each of which is independently unmodified or chemically modified
  • [N] represents nucleotides that are ribonucleotides
  • XI and X2 are independently integers from 0 to 4
  • X3 is an integer from 15 to 30
  • X4 is an integer from 9 to 30;
  • X5 is an integer from 0 to 6, provided that the sum of X4 and X5 is 15-30.
  • nucleotides of the at least a 15 nucleotide sequence of SEQ ID NO: 1049, SEQ ID NO: 1091, SEQ ID NO: 1099, or SEQ ID NO: 1101 form a contiguous stretch of nucleotides.
  • the siNA molecule of formula A can contain one or more nucleotide deletions, substitutions, mismatches and/or additions to the at least 15 nucleotide sequence of SEQ ID NO: 1049, SEQ ID NO: 1091, SEQ ID NO: 1099, or SEQ ID NO: 1101; provided, however, that the siNA molecule maintains its activity, for example, to mediate RNAi.
  • deletion, substitution, mismatch and/or addition can result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair.
  • the invention features a double- stranded short interfering nucleic acid (siNA) of formula (A); wherein
  • one or more pyrimidine nucleotides in ⁇ 4 positions are independently 2'-deoxy- 2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof;
  • one or more purine nucleotides in ⁇ 4 positions are independently 2'-deoxy-2'- fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof;
  • one or more pyrimidine nucleotides in ⁇ 3 positions are independently 2'-deoxy- 2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof;
  • one or more purine nucleotides in ⁇ 3 positions are independently 2'-deoxy-2'- fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof.
  • the invention features a double-stranded short interfering nucleic acid (siNA) molecule of formula (A); wherein
  • the invention features a double-stranded short interfering nucleic acid (siNA) molecule of formula (A); wherein SIR-ONC-00001
  • the invention features a double-stranded short interfering nucleic acid (siNA) molecule of formula (A); wherein
  • the invention features a double-stranded short interfering nucleic acid (siNA) molecule of formula (A) further comprising one or more phosphorothioate internucleotide linkages.
  • siNA short interfering nucleic acid
  • siNA molecules having formula A comprise a terminal phosphate group at the 5 '-end of the antisense strand or antisense region of the nucleic acid molecule.
  • siNA molecules having formula A comprise caps (B) at the 3' and 5' ends of the sense strand or sense region.
  • siNA molecules having formula A comprise caps (B) at the 3 '-end of the antisense strand or antisense region.
  • siNA molecules having formula A comprise caps (B) at the 3' and 5' ends of the sense strand or sense region and caps (B) at the 3 '-end of the antisense strand or antisense region.
  • siNA molecules having formula A comprise caps (B) only at the 5'-end of the sense (upper) strand of the double- stranded nucleic acid molecule.
  • siNA molecules having formula A further comprise one or more phosphorothioate intemucleotide linkages between the nucleotides.
  • siNA molecules having formula A comprise one or more phosphorothioate intemucleotide linkages between the first terminal (N) and the adjacent nucleotide on the 3 'end of the sense strand, antisense strand, or both sense strand and antisense strands of the nucleic acid molecule.
  • one or more of the nucleotides of siNA molecules having formula A have a universal base.
  • siNA molecules having formula A have at position 14 from the 5'-end of the antisense strand a ribonucleotide when the nucleotide at that position 14 is a purine. In other embodiments, siNA molecules having formula A have at position 14 from the 5'-end of the antisense strand a ribonucleotide, a 2'-deoxy-2'-fluoro nucleotide or a 2'-0-methyl nucleotide when the nucleotide at that position 14 is a pyrimidine nucleotide.
  • siNA molecules having formula A comprises (N) nucleotides in the antisense strand (lower strand) that are complementary to nucleotides in a MET target polynucleotide sequence, which also has complementarity to the N and [N] nucleotides of the antisense (lower) strand.
  • siNAs of the invention can be obtained using a number of techniques known to those of skill in the art.
  • the siNA can be chemically synthesized or may be encoded by plasmid (e.g., transcribed as sequences that automatically fold into duplexes with hairpin loops.).
  • siNA can also be generated by cleavage of longer dsRNA (e.g., dsRNA greater than about 25 nucleotides in length) by the E coli RNase II or Dicer. These enzymes process the dsRNA into biologically active siNA (see, e.g., Yang et al, PNAS USA 99:9942-9947 (2002); Calegari et al.
  • siNA of the invention are chemically synthesized.
  • Oligonucleotides e.g., certain modified oligonucleotides or portions of oligonucleotides lacking ribonucleotides
  • SIR-ONC-00001 synthesized using protocols known in the art, for example as described in Caruthers et al, 1992, Methods in Enzymology 211, 3-19, Thompson et al., International PCT Publication No. WO 99/54459, Wincott et al, 1995, Nucleic Acids Res. 23, 2677-2684, Wincott et al, 1997, Methods Mol.
  • oligonucleotides makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end.
  • siNA molecules without modifications are synthesized using procedures as described in Usman et al, 1987, J. Am. Chem. Soc, 109, 7845; Scaringe et al, 1990, Nucleic Acids Res., 18, 5433. These syntheses makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end that can be used for certain siNA molecules of the invention.
  • the siNA molecules of the invention are synthesized, deprotected, and analyzed according to methods described in U.S. Patent Nos. 6,995,259, 6,686,463, 6,673,918, 6,649,751, 6,989,442, and U.S. Patent Application No. 10/190,359.
  • the siNA molecules of the present invention can be synthesized separately and joined together post-synthetically, for example, by ligation (Moore et al, 1992, Science 256, 9923; Draper et al, International PCT Publication No. WO 93/23569; Shabarova et al, 1991, Nucleic Acids Research 19, 4247; Bellon et al, 1997, Nucleosides & Nucleotides, 16, 951; Bellon et al, 1997, Bioconjugate Chem. 8, 204), or by hybridization following synthesis and/or deprotection.
  • siNA molecules of the invention can also be synthesized using the teachings of Scaringe et al, US Patent Nos. 5,889,136; 6,008,400; and 6,111,086.
  • siNA molecules of the invention that interact with and down-regulate gene encoding target MET molecules can be expressed and delivered from transcription units (see for example Couture et al, 1996, TIG., 12, 510) inserted into DNA or RNA vectors.
  • the recombinant vectors can be DNA plasmids or viral vectors.
  • siNA expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus, retrovirus, adenovirus, or alphavirus.
  • pol III based constructs are used to express nucleic acid molecules of the invention. Transcription of the siNA molecule sequences can be driven from a promoter for eukaryotic RNA polymerase I (pol I), RNA polymerase II (pol II), or RNA polymerase III (pol III), (see for example, Thompson, U.S. Patent. Nos. 5,902,880 and 6,146,886). (See also, Izant and Weintraub, 1985, Science, 229, 345; McGarry and Lindquist, 1986, Proc. Natl. Acad. Set, USA 83, 399; Scanlon et al, 1991, Proc. Natl. Acad. Sci.
  • Transcripts from pol II or pol III promoters are expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type depends on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby.
  • Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Elroy-Stein and Moss, 1990, Proc. Natl. Acad. Sci. U S A, 87, 6743-7; Gao and Huang 1993, Nucleic Acids Res., 21, 2867-72; Lieber et al, 1993, Methods Enzymol, 217, 47-66; Zhou et al, 1990, Mol. Cell. Biol, 10, 4529-37).
  • nucleic acid molecules expressed from such promoters can function in mammalian cells ⁇ e.g. Kashani-Sabet et al, 1992, Antisense Res.
  • transcription units such as the ones derived from genes encoding U6 small nuclear (snRNA), transfer RNA (tRNA) and adenovirus VA RNA are useful in generating high SIR-ONC-00001 concentrations of desired RNA molecules such as siNA in cells (Thompson et al., supra; Couture and Stinchcomb, 1996, supra; Noonberg et al., 1994, Nucleic Acid Res., 22, 2830; Noonberg et al., U.S. Pat. No. 5,624,803; Good et al., 1997, Gene Ther., 4, 45; Beigelman et al., International PCT Publication No. WO 96/18736.
  • siNA transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated virus vectors), or viral RNA vectors (such as retroviral or alphavirus vectors) (for a review see Couture and Stinchcomb, 1996, supra).
  • plasmid DNA vectors such as adenovirus or adeno-associated virus vectors
  • viral RNA vectors such as retroviral or alphavirus vectors
  • Vectors used to express the siNA molecules of the invention can encode one or both strands of an siNA duplex, or a single self-complementary strand that self hybridizes into an siNA duplex.
  • the nucleic acid sequences encoding the siNA molecules of the instant invention can be operably linked in a manner that allows expression of the siNA molecule (see for example Paul et al., 2002, Nature Biotechnology, 19, 505; Miyagishi and Taira, 2002, Nature Biotechnology, 19, 497; Lee et al, 2002, Nature Biotechnology, 19, 500; and Novina et al, 2002, Nature Medicine, advance online publication doi: 10.1038/nm725).
  • siNA molecules of the invention are added directly, or can be complexed with cationic lipids, packaged within liposomes, or as a recombinant plasmid or viral vectors which express the siNA molecules, or otherwise delivered to target cells or tissues.
  • Methods for the delivery of nucleic acid molecules are described in Akhtar et al., 1992, Trends Cell Bio., 2, 139; Delivery Strategies for Antisense Oligonucleotide Therapeutics, ed. Akhtar, 1995, Maurer et al., 1999, Mol. Membr. Biol., 16, 129-140; Hofland and Huang, 1999, Handb. Exp.
  • Nucleic acid molecules can be administered to cells by a variety of methods known to those of skill in the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as biodegradable polymers, hydrogels, cyclodextrins (see for example, Gonzalez et al., 1999, Bioconjugate Chem., SIR-ONC-00001
  • the present invention provides carrier systems containing the siNA molecules described herein.
  • the carrier system is a lipid-based carrier system, cationic lipid, or liposome nucleic acid complexes, a liposome, a micelle, a virosome, a lipid nanoparticle or a mixture thereof.
  • the carrier system is a polymer- based carrier system such as a cationic polymer-nucleic acid complex.
  • the carrier system is a cyclodextrin-based carrier system such as a cyclodextrin polymer-nucleic acid complex.
  • the carrier system is a protein-based carrier system such as a cationic peptide-nucleic acid complex.
  • the carrier system in a lipid nanoparticle ("LNP") formulation.
  • the siNA molecules of the invention are formulated with a lipid nanoparticle composition such as is described in U.S. Patent Application Nos. 11/353,630, 11/586,102, 61/189,295, 61/204,878, 61/235,476, 61/249,807, and 61/298,022.
  • the siNA molecules of the invention are formulated with a lipid nanoparticle composition comprising a cationic lipid/Cholesterol/PEG-C-DMA/DSPC in a 40/48/2/10 ratio or a cationic lipid/Cholesterol/PEG-DMG/DSPC in a 40/48/2/10 ratio.
  • the cationic lipid is DLinDMA (see Table 14) and the PEG is PEG- DMG.
  • the N/P ratio of the formulation is 2.8.
  • lipid nanoparticle formulations described in Table 13 are applied to any siNA molecule or combination of siNA molecules herein.
  • the invention features a composition comprising an siNA molecule of the invention formulated as any of formulation LNP-051; LNP-053; LNP-054; LNP-069; LNP-073; LNP-077; LNP-080; LNP-082; LNP-083; LNP-060; LNP-061; LNP-086; LNP-097; LNP-098; LNP-099; LNP-100; LNP-101; LNP-102; LNP-103; or LNP-104 (see Table 13).
  • the invention features a composition comprising an siNA molecule of the invention formulated with any of the cationic lipid formulations described in U.S. Patent Application Nos. 61/189,295, 61/204,878, 61/235,476, 61/249,807, and 61/298,022.
  • the invention features conjugates and/or complexes of siNA molecules of the invention.
  • Such conjugates and/or complexes can be used to facilitate delivery of siNA molecules into a biological system, such as a cell.
  • the conjugates and complexes provided by the instant invention can impart therapeutic activity by transferring therapeutic compounds across cellular membranes, altering the pharmacokinetics, and/or modulating the localization of nucleic acid molecules of the invention.
  • Non-limiting, examples of such conjugates are described in U.S. Publication Nos. US2008/0152661 Al and US 2004/0162260 Al (e.g., CDM-LBA, CDM-Pip-LBA, CDM-PEG, CDM-NAG, etc.) and U.S. Patent Application Nos. 10/427,160 and 10/201,394; and U.S. Patent Nos. 6,528,631; 6,335,434; 6,235,886; 6,153,737; 5,214,136; and 5,138,045.
  • polyethylene glycol can be covalently attached to siNA compounds of the present invention.
  • the attached PEG can be any molecular weight, preferably from about 100 to about 50,000 daltons (Da).
  • the invention features compositions or formulations comprising surface-modified liposomes containing poly (ethylene glycol) lipids (PEG-modified, or long-circulating liposomes or stealth liposomes) and siNA molecules of the invention, such as is disclosed in for example, International PCT Publication No. WO 96/10391; Ansell et al., International PCT Publication No. WO 96/10390; Holland et al, International PCT Publication No. WO 96/10392.
  • the siNA molecules of the invention can also be formulated or complexed with polyethyleneimine and derivatives thereof, such as polyethyleneimine- polyethyleneglycol-N-acetylgalactosamine (PEI-PEG-GAL) or polyethyleneimine- polyethyleneglycol-tri-N-acetylgalactosamine (PEI-PEG-triGAL) derivatives.
  • polyethyleneimine- polyethyleneglycol-N-acetylgalactosamine PEI-PEG-GAL
  • PEI-PEG-triGAL polyethyleneimine- polyethyleneglycol-tri-N-acetylgalactosamine
  • siNA molecules of the invention are complexed with membrane disruptive agents such as those described in U.S. Patent Application Publication No. 20010007666.
  • the membrane disruptive agent or agents and the siNA molecule are also complexed with a cationic lipid or helper lipid molecule, such as those lipids described in U.S. Patent No. 6,235,310.
  • siNA molecules of the invention are complexed with delivery systems as described in U.S. Patent Application Publication Nos. 2003077829; 20050287551 ; 20050164220; 20050191627; 20050118594; 20050153919; 20050085486; and 20030158133; and International PCT Publication Nos. WO 00/03683 and WO 02/087541.
  • a liposomal formulation of the invention comprises an siNA molecule of the invention (e.g., siNA) formulated or complexed with compounds and compositions described in U.S. Patent Nos. 6,858,224; 6,534,484; 6,287,591 ; 6,835,395; 6,586,410; 6,858,225; 6,815,432; 6,586,001 ; 6,120,798; 6,977,223; 6,998, 115; 5,981,501 ; 5,976,567; 5,705,385; and U.S. Patent Application Publication Nos.
  • siNA siNA
  • siNA molecule expressing vectors can be systemic, such as by intravenous or intra-muscular administration, by administration to target cells ex-planted from a subject followed by reintroduction into the subject, or by any other means that would allow for introduction into the desired target cell (for a review see Couture et ah, 1996, TIG., 12, 510).
  • Such recombinant plasmids can also be administered directly or in conjunction with a suitable delivery reagents, including, for example, the Minis Transit LT1 lipophilic reagent; lipofectin; lipofectamine; cellfectin; polycations (e.g., polylysine) or liposomes lipid-based carrier system, cationic lipid, or liposome nucleic acid complexes, a micelle, a virosome, a lipid nanoparticle.
  • a suitable delivery reagents including, for example, the Minis Transit LT1 lipophilic reagent; lipofectin; lipofectamine; cellfectin; polycations (e.g., polylysine) or liposomes lipid-based carrier system, cationic lipid, or liposome nucleic acid complexes, a micelle, a virosome, a lipid nanoparticle.
  • the present invention also provides nucleic acids in kit form.
  • the kit may comprise a container.
  • the kit typically contains a nucleic acid of the invention with instructions for its administration.
  • the nucleic acids may have a targeting moiety attached. Methods of attaching targeting moieties (e.g. antibodies, proteins) are known to those of skill in the art.
  • the nucleic acids are chemically modified.
  • the kit contains more than one siNA molecule of the invention.
  • the kits may comprise an siNA molecule of the invention with a pharmaceutically acceptable carrier or diluent.
  • the kits may further comprise excipients.
  • nucleic acid molecules of the present invention can be used in assays to diagnose disease state related of MET levels.
  • nucleic acid molecules and pharmaceutical compositions can be used to treat disease states related to MET RNA levels.
  • cancers including solid tumors such as skin, breast, brain, cervical carcinomas, testicular carcinomas, etc.
  • Non-limiting examples of such cancers include: Cardiac: sarcoma fibrosarcoma, rhabdomyosarcoma, and liposarcoma; Lun : bronchogenic carcinoma, squamous cell carcinoma, undifferentiated small cell carcinoma, undifferentiated large cell carcinoma, lung adenocarcinoma, alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous harmartoma, mesothelioma, non-small cell lung carcinoma, small cell lung cancer, and lung carcinoma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma), stomach (carcinoma, leiomyosarcoma), pan
  • the siNA molecules of the invention can degrade the target MET mRNA (and thus inhibit the diseases stated above). Inhibition of a disease can be evaluated by directly measuring the progress of the disease in a subject. It can also be inferred through observing a change or reversal in a condition associated with the disease. Additionally, the siNA molecules of the invention can be used as a prophylaxis. Thus, the use of the nucleic acid molecules and pharmaceutical compositions of the invention can be used to ameliorate, treat, prevent, and/or cure these diseases and others associated with regulation of MET gene expression. SIR-ONC-00001
  • siNA molecules of the instant invention provide useful reagents and methods for a variety of therapeutic, prophylactic, cosmetic, veterinary, diagnostic, target validation, genomic discovery, genetic engineering, and pharmacogenomic applications. a. Formulations
  • compositions of the siNA molecules described i.e., compositions in a pharmaceutically acceptable carrier or diluent.
  • pharmaceutical compositions include salts of the above compounds, e.g., acid addition salts, for example, salts of hydrochloric, hydrobromic, hydroiodic, acetic acid, and benzene sulfonic acid.
  • Other salts include for example, sodium, potassium, manganese, ammonium, and calcium salts.
  • formulations or compositions can comprise a pharmaceutically acceptable carrier or diluent as is generally known in the art.
  • the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1.
  • the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1049.
  • the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 43.
  • the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1091.
  • the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 51. In another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1099. In another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 53. In yet another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1101. In still another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising formula (A). SIR-ONC-00001
  • the siNA molecules of the invention are preferably formulated as pharmaceutical compositions prior to administering to a subject, according to techniques known in the art.
  • Pharmaceutical compositions of the present invention are characterized as being at least sterile and pyrogen-free. Methods for preparing pharmaceutical composition of the invention are within the skill in the art for example as described in Remington's Pharmaceutical Science, 17 th ed., Mack Publishing Company, Easton, Pa. (1985).
  • compositions of the invention further comprise conventional pharmaceutical excipients and/or additives.
  • suitable pharmaceutical excipients include preservatives, flavoring agents, stabilizers, antioxidants, osmolality adjusting agents, buffers, and pH adjusting agents.
  • Suitable additives include physiologically biocompatible buffers (e.g., trimethylamine hydrochloride), addition of chelants (such as, for example, DTPA or DTPA-bisamide) or calcium chelate complexes (as for example calcium DTPA, CaNaDTPA-bisamide), or, optionally, additions of calcium or sodium salts (for example, calcium chloride, calcium ascorbate, calcium gluconate or calcium lactate).
  • chelants such as, for example, DTPA or DTPA-bisamide
  • calcium chelate complexes as for example calcium DTPA, CaNaDTPA-bisamide
  • calcium or sodium salts for example, calcium chloride, calcium ascorbate, calcium gluconate or calcium lactate.
  • antioxidants and suspending agents can be used.
  • Non-limiting examples of various types of formulations for local administration include ointments, lotions, creams, gels, foams, preparations for delivery by transdermal patches, powders, sprays, aerosols, capsules or cartridges for use in an inhaler or insufflator or drops (for example eye or nose drops), solutions/suspensions for nebulization, suppositories, pessaries, retention enemas and chewable or suckable tablets or pellets (for example for the treatment of aphthous ulcers) or liposome or microencapsulation preparations.
  • Ointments, creams and gels can, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agent and/or solvents.
  • suitable thickening and/or gelling agent and/or solvents can thus, for example, include water and/or an oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil, or a solvent such as polyethylene glycol. Thickening agents and gelling agents which can be used according to the nature of the base.
  • Non-limiting examples of such agents include soft paraffin, aluminum stearate, cetostearyl alcohol, polyethylene glycols, woolfat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents.
  • Soft paraffin aluminum stearate, cetostearyl alcohol, polyethylene glycols, woolfat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents.
  • lotions can be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents or thickening agents.
  • powders for external application can be formed with the aid of any suitable powder base, for example, talc, lactose or starch.
  • Drops can be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilizing agents, suspending agents or preservatives.
  • compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients can be, for example, inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets can be uncoated or they can be coated by known techniques. In some cases such coatings can be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monosterate or glyceryl distearate can be employed.
  • Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active materials in a mixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents can SIR-ONC-00001 be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate; or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example
  • the aqueous suspensions can also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavoring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • sweetening agents such as sucrose or saccharin.
  • Oily suspensions can be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions can contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents and flavoring agents can be added to provide palatable oral preparations.
  • These compositions can be preserved by the addition of an antioxidant such as ascorbic acid
  • compositions of the invention can also be in the form of oil-in-water emulsions.
  • the oily phase can be a vegetable oil or a mineral oil or mixtures of these.
  • Suitable emulsifying agents can be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions can also contain sweetening and flavoring agents.
  • Syrups and elixirs can be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations can also contain a demulcent, a preservative and flavoring and coloring agents.
  • the pharmaceutical compositions can be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above.
  • the sterile injectable preparation can also SIR-ONC-00001 be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • a non-toxic parentally acceptable diluent or solvent for example as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono-or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the nucleic acid molecules of the invention can also be administered in the form of suppositories, e.g., for rectal administration of the drug.
  • suppositories e.g., for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials include cocoa butter and polyethylene glycols.
  • Nucleic acid molecules of the invention can be administered parenterally in a sterile medium.
  • the drug depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle.
  • adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle.
  • the siNA and LNP compositions and formulations provided herein for use in pulmonary delivery further comprise one or more surfactants.
  • Suitable surfactants or surfactant components for enhancing the uptake of the compositions of the invention include synthetic and natural as well as full and truncated forms of surfactant protein A, surfactant protein B, surfactant protein C, surfactant protein D and surfactant Protein E, di- saturated phosphatidylcholine (other than dipalmitoyl), dipalmitoylphosphatidylcholine, phosphatidylcholine, phosphatidylglycerol, phosphatidylinositol, phosphatidylethanolamine, phosphatidylserine; phosphatidic acid, ubiquinones, lysophosphatidylethanolamine, lysophosphatidylcholine, palmitoyl-lysophosphatidylcholine, dehydroepiandrosterone,
  • siNAs and pharmaceutical formulations according to the invention can be administered to a subject alone or used in combination with or include one or more other therapeutic agents, for example, anticancer agents.
  • anticancer agents for example, anticancer agents.
  • combinations of the presently disclosed compounds with other anti-cancer or chemotherapeutic agents are within the scope of the invention. Examples of such agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S. Hellman (editors), 6 th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved.
  • Such anti-cancer agents include, but are not limited to, the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic/cytostatic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG- CoA reductase inhibitors and other angiogenesis inhibitors, inhibitors of cell proliferation and survival signaling, apoptosis inducing agents and agents that interfere with cell cycle checkpoints.
  • the siNAs of the invention are also useful in combination with any therapeutic agent used in the treatment of HCC, for example, but not limitation sorafenib.
  • the instant compounds are particularly useful when co-administered with radiation therapy.
  • the invention provides a combination comprising an siNA molecule of the invention, such as for example, but not limitation, an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A) or a pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof together with one or more anti-cancer or chemotherapeutic agents.
  • an siNA molecule of the invention such as for example, but not limitation, an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A) or a pharmaceutically acceptable salt
  • the instant siNA molecules of the invention are useful in combination with known anti-cancer agents including the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors, HIV protease inhibitors, reverse transcriptase inhibitors, and other angiogenesis inhibitors.
  • known anti-cancer agents including the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors, HIV protease inhibitors, reverse transcriptase inhibitors, and other angiogenesis inhibitors.
  • estrogen receptor modulators examples include, but are not limited to, tamoxifen, raloxifene, idoxifene, LY353381, LY117081, toremifene, fulvestrant, 4-[7-(2,2-dimethyl-l-oxopropoxy-4-methyl-2- [4- [2- ( 1 -piperidinyl)ethoxy] phenyl] -2H- 1 -benzopyran-3-yl] -phenyl-2,2-dimethylpropanoate, 4,4' -dihydroxybenzophenone-2,4-dinitrophenyl-hydrazone, and SH646.
  • Examples of androgen receptor modulators that can be used in combination with the compounds of the invention include, but are not limited to, finasteride and other 5a-reductase inhibitors, nilutamide, flutamide, bicalutamide, liarozole, and abiraterone acetate.
  • retinoid receptor modulators examples include, but are not limited to, bexarotene, tretinoin, 13-cis- retinoic acid, 9-cis-retinoic acid, -difluoromethylornithine, ILX23-7553, trans-N-(4'- hydroxyphenyl) retinamide, and N-4-carboxyphenyl retinamide.
  • cytotoxic agents that can be used in combination with the compounds of the invention include, but are not limited to, sertenef, cachectin, ifosfamide, tasonermin, lonidamine, carboplatin, altretamine, prednimustine, dibromodulcitol, ranimustine, fotemustine, nedaplatin, oxaliplatin, temozolomide, heptaplatin, estramustine, improsulfan tosilate, trofosfamide, nimustine, dibrospidium chloride, pumitepa, lobaplatin, satraplatin, profiromycin, cisplatin, irofulven, dexifosfamide, cis-aminedichloro(2-methyl-pyridine)platinum, benzylguanine, glufosfamide, GPX100, (trans, trans, trans)-bis-mu
  • hypoxia activatable compound that can be used in combination with the compounds of the invention is tirapazamine.
  • proteasome inhibitors examples include, but are not limited to, lactacystin and bortezomib.
  • microtubule inhibitors/microtubule-stabilising agents that can be used in combination with the compounds of the invention include, but are not limited to, paclitaxel, vindesine sulfate, 3',4'-didehydro-4'-deoxy-8'-norvincaleukoblastine, docetaxol, rhizoxin, dolastatin, mivobulin isethionate, auristatin, cemadotin, RPR109881, BMS 184476, vinflunine, cryptophycin, 2,3,4,5, 6-pentafluoro-N-(3-fluoro-4-methoxyphenyl) benzene sulfonamide, anhydrovinblastine, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-prolyl-L-proline-t- butylamide, TDX258, the epothilones (see
  • topoisomerase inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, are topotecan, hycaptamine, irinotecan, rubitecan, 6-ethoxypropionyl-3',4'-0-exo-benzylidene-chartreusin, 9-methoxy-N,N- dimethyl-5-nitropyrazolo[3,4,5-kl]acridine-2-(6H) propanamine, l-amino-9-ethyl-5-fluoro-2,3- dihydro-9 -hydroxy-4-methyl- 1 H, 12H-benzo [de]pyrano [3 ' ,4' :b,7] -indolizino [ 1 ,2b] quinoline- 10,13(9H,15H)dione, lurtotecan, 7-[2-(N-isopropylamino)ethyl]-(20S)camptothec
  • inhibitors of mitotic kinesins include, but are not limited to, inhibitors described in PCT Publications WO 01/30768, WO 01/98278, WO 03/050,064, WO 03/050,122, WO 03/049,527, WO 03/049,679, WO 03/049,678, WO04/039774, WO03/079973, WO03/099211, WO03/105855, WO03/106417, WO04/037171, WO04/058148, WO04/058700, WO04/126699, WO05/018638, WO05/019206, WO05/019205, WO05/018547, WO05/017190, US2005/0176776.
  • inhibitors of mitotic kinesins include, but are not limited to inhibitors of KSP, inhibitors of MKLPl, inhibitors of CENP-E, inhibitors of MCAK, inhibitors of Kifl4, inhibitors of Mphosphl and inhibitors of Rab6-KIFL.
  • histone deacetylase inhibitors examples include, but are not limited to, TSA, oxamflatin, PXD101, MG98, valproic acid and scriptaid. Further reference to other histone deacetylase inhibitors may be found in the following manuscript; Miller, T.A. et al. J. Med. Chem. 46(24):5097-5116 (2003).
  • Inhibitors of kinases involved in mitotic progression include, but are not limited to, inhibitors of aurora kinase, inhibitors of Polo-like kinases (PLK) (in particular inhibitors of PLK-1), inhibitors of bub-1 and inhibitors of bub-Rl.
  • PLK Polo-like kinases
  • Antiproliferative agents that can be used in combination with the compounds of the invention include, but are not limited to, antisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231, and INX3001, and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fludarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2'-deoxy-2'-methylidenecytidine, 2'- fluoromethylene-2'-deoxycytidine, N-[5-(2,3-dihydro-benzofuryl)sulf
  • Examples of monoclonal antibody targeted therapeutic agents that can be used in combination with the compounds of the invention include those therapeutic agents which have cytotoxic agents or radioisotopes attached to a cancer cell specific or target cell specific monoclonal antibody, such as, for example, Bexxar.
  • HMG-CoA reductase inhibitors that may be used that can be used in combination with the compounds of the invention include, but are not limited to, lovastatin
  • prenyl-protein transferase inhibitors examples include, but are not limited to, can be found in the following publications and patents: WO 96/30343, WO 97/18813, WO 97/21701, WO 97/23478, WO 97/38665, WO 98/28980, WO 98/29119, WO 95/32987, U.S. Pat. No. 5,420,245, U.S. Pat. No. 5,523,430, U.S. Pat. No. 5,532,359, U.S. Pat. No. 5,510,510, U.S. Pat. No. 5,589,485, U.S. Pat. No.
  • angiogenesis inhibitors examples include, but are not limited to, tyrosine kinase inhibitors, such as inhibitors of the tyrosine kinase receptors Flt-1 (VEGFR1) and Flk-l/KDR (VEGFR2), inhibitors of epidermal-derived, fibroblast-derived, or platelet derived growth factors, MMP (matrix metalloprotease) inhibitors, integrin blockers, interferon- , interleukin-12, pentosan polysulfate, cyclooxygenase inhibitors, including nonsteroidal anti-inflammatories (NSAIDs) like aspirin and ibuprofen as well as selective cyclooxy-genase-2 inhibitors like celecoxib and rofecoxib (PNAS, Vol.
  • NSAIDs nonsteroidal anti-inflammatories
  • steroidal anti-inflammatories such as corticosteroids, mineralocorticoids, dexamethasone, prednisone, prednisolone, methylpred, betamethasone), carboxyamidotriazole, combretastatin A-4, squalamine, 6-0- chloroacetyl-carbonyl)-fumagillol, thalidomide, angiostatin, troponin- 1, angiotensin II antagonists (see Fernandez et al., J. Lab. Clin. Med.
  • agents that modulate or inhibit angiogenesis may also be used in combination with the compounds of the instant inventionand include agents that modulate or inhibit the coagulation and fibrinolysis systems (see review in Clin. Chem. La. Med. 38:679-692 (2000)).
  • agents that modulate or inhibit the coagulation and fibrinolysis pathways that can be used in combination with the compounds of the invention include, but are SIR-ONC-00001 not limited to, heparin (see Thromb. Haemost. 80: 10-23 (1998)), low molecular weight heparins and carboxypeptidase U inhibitors (also known as inhibitors of active thrombin activatable fibrinolysis inhibitor [TAFIa]) (see Thrombosis Res. 101:329-354 (2001)).
  • TAFIa inhibitors have been described in PCT Publication WO 03/013,526 and U,S, Ser. No. 60/349,925 (filed January 18, 2002).
  • Agents that interfere with cell cycle checkpoints that can be used in combination with the compounds of the invention include, but are not limited to, inhibitors of ATR, ATM, the Chkl and Chk2 kinases and cdk and cdc kinase inhibitors and are specifically exemplified by 7- hydroxystaurosporin, flavopiridol, CYC202 (Cyclacel) and BMS-387032.
  • RTKs receptor tyrosine kinases
  • agents that interfere with receptor tyrosine kinases (RTKs) include, but are not limited to, inhibitors of c- Kit, Eph, PDGF, Flt3 and MET. Further agents include inhibitors of RTKs as described by Bume- Jensen and Hunter, Nature, 411:355-365, 2001.
  • Inhibitors of cell proliferation and survival signaling pathway that can be used in combination with the compounds of the invention include, but are not limited to, inhibitors of EGFR (for example gefitinib and erlotinib), inhibitors of ERB-2 (for example trastuzumab), inhibitors of IGFR, inhibitors of cytokine receptors, inhibitors of MET, inhibitors of PI3K (for example LY294002), serine/threonine kinases (including but not limited to inhibitors of Akt such as described in WO 02/083064, WO 02/083139, WO 02/083140, US 2004-0116432, WO 02/083138, US 2004-0102360, WO 03/086404, WO 03/086279, WO 03/086394, WO 03/084473, WO 03/086403, WO 2004/041162, WO 2004/096131, WO 2004/096129, WO 2004/096135, WO 2004/09
  • Apoptosis inducing agents that can be used in combination with the compounds of the invention include, but are not limited to, activators of TNF receptor family members (including the TRAIL receptors). SIR-ONC-00001
  • NSAIDs that are selective COX-2 inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, those NSAIDs disclosed in U.S. Pat. 5,474,995, U.S. Pat. 5,861,419, U.S. Pat. 6,001,843, U.S. Pat. 6,020,343, U.S. Pat. 5,409,944, U.S. Pat. 5,436,265, U.S. Pat. 5,536,752, U.S. Pat. 5,550, 142, U.S. Pat. 5,604,260, U.S. 5,698,584, U.S. Pat. 5,710,140, WO 94/15932, U.S. Pat. 5,344,991, U.S. Pat.
  • Inhibitors of COX-2 that are particularly useful in combination with the compounds of the invention include: 3-phenyl-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; and 5-chloro- 3-(4-methylsulfonyl)-phenyl-2-(2-methyl-5-pyridinyl)pyridine; or a pharmaceutically acceptable salt thereof.
  • Angiogenesis inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, endostatin, ukrain, ranpirnase, IM862, 5-methoxy-4-[2- methyl-3-(3-methyl-2-butenyl)oxiranyl]- l-oxaspiro[2,5]oct-6-yl(chloroacetyl)carbamate, acetyldinanaline, 5-amino- l-[[3,5-dichloro-4-(4-chlorobenzoyl)-phenyl]methyl]-lH- l,2,3- triazole-4-carboxamide,CM101, squalamine, combretastatin, RPI4610, NX31838, sulfated mannopentaose phosphate, 7,7-(carbonyl-bis[imino-N-methyl-4,2-pyrrolocarbonylimino[N- methyl-4,
  • Tyrosine kinase inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, N-(trifluoromethylphenyl)-5-methylisoxazol-4- carboxamide, 3-[(2,4-dimethylpyrrol-5-yl)methylidenyl)indolin-2-one, 17-(allylamino)- 17- demethoxygeldanamycin, 4-(3-chloro-4-fluorophenylamino)-7-methoxy-6-[3-(4- morpholinyl)propoxyl]quinazoline, N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4- quinazolinamine, BIBX1382, 2,3,9, 10,11, 12-hexahydro- 10-(hydroxymethyl)- 10-hydroxy-9- methyl-9,12-epoxy- lH-diindolo[l,2,3-fg:3'
  • Combinations with compounds other than anti-cancer compounds are also encompassed in the instant compositions and methods.
  • combinations of the instantly claimed compounds with PPAR- ⁇ (i.e., PPAR-gamma) agonists and PPAR- ⁇ (i.e., PPAR-delta) agonists are useful in the treatment of certain malingnancies.
  • PPAR-y and PPAR- ⁇ are the nuclear peroxisome proliferator-activated receptors ⁇ and ⁇ .
  • the expression of PPAR- ⁇ on endothelial cells and its involvement in angiogenesis has been reported in the literature (see J. Cardiovasc. Pharmacol. 1998; 31:909-913; J. Biol. Chem.
  • Examples of PPAR- ⁇ agonists and PPAR- ⁇ / agonists that can be used in combination with the compounds of the invention include, but are not limited to, thiazolidinediones (such as DRF2725, CS-011, troglitazone, rosiglitazone, and pioglitazone), fenofibrate, gemfibrozil, clofibrate, GW2570, SB219994, AR-H039242, JTT-501, MCC-555, GW2331, GW409544, NN2344, KRP297, NP0110, DRF4158, NN622, GI262570, PNU182716, DRF552926, 2-[(5,7-dipropyl-3-trifluoromethyl-l,2-benzisoxazol-6-yl)oxy]-2-methylpropionic acid (disclosed in USSN 09/782,856), and 2(R)-7-(3-(2-chloro-4
  • Another embodiment of the instant invention is the use of the presently disclosed compounds in combination with gene therapy for the treatment of cancer.
  • Gene therapy can be used to deliver any tumor suppressing gene. Examples of such genes include, but are not limited to, p53, which can be delivered via recombinant virus -mediated gene transfer (see U.S. Pat. No.
  • a uPA/uPAR antagonist (Adenovirus-Mediated Delivery of a SIR-ONC-00001 uPA/uPAR Antagonist Suppresses Angiogenesis-Dependent Tumor Growth and Dissemination in Mice," Gene Therapy, August 1998;5(8): 1105-13), and interferon gamma (/ Immunol 2000;164:217-222).
  • the compounds of the instant invention may also be administered in combination with an inhibitor of inherent multidrug resistance (MDR), in particular MDR associated with high levels of expression of transporter proteins.
  • MDR inhibitors include inhibitors of p- glycoprotein (P-gp), such as LY335979, XR9576, OC144-093, R101922, VX853 and PSC833 (valspodar).
  • a compound of the present invention may be employed in conjunction with antiemetic agents to treat nausea or emesis, including acute, delayed, late -phase, and anticipatory emesis, which may result from the use of a compound of the present invention, alone or with radiation therapy.
  • a compound of the present invention may be used in conjunction with other anti-emetic agents, especially neurokinin- 1 receptor antagonists, 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, a corticosteroid such as Decadron (dexamethasone), Kenalog, Aristocort, Nasalide, Preferid, Benecorten or others such as disclosed in U.S.Patent Nos.
  • neurokinin- 1 receptor antagonists especially 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, a corticosteroid such as Decadron (dexamethasone), Kenalog, Aristocort, Nasalide, Preferid, Benecorten or others such as disclosed in U.S.Patent No
  • an antidopaminergic such as the phenothiazines (for example prochlorperazine, fluphenazine, thioridazine and mesoridazine), metoclopramide or dronabinol.
  • an anti-emesis agent selected from a neurokinin- 1 receptor antagonist, a 5HT3 receptor antagonist and a corticosteroid is administered as an adjuvant for the treatment or prevention of emesis that may result upon administration of the instant compounds.
  • Neurokinin- 1 receptor antagonists of use in conjunction with the compounds of the present invention are fully described, for example, in U.S. Pat. Nos. 5,162,339, 5,232,929, 5,242,930, 5,373,003, 5,387,595, 5,459,270, 5,494,926, 5,496,833, 5,637,699, 5,719,147; European Patent Publication Nos.
  • the neurokinin- 1 receptor antagonist for use in conjunction with the compounds of the present invention is selected from: 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)- phenyl)ethoxy)-3 -(S )- (4-fluorophenyl)-4- (3- (5 -oxo- 1 H,4H- 1 ,2,4-triazolo)methyl)morpholine, or a pharmaceutically acceptable salt thereof, which is described in U.S. Pat. No. 5,719,147.
  • a compound of the instant invention may also be useful for treating or preventing cancer, including bone cancer, in combination with bisphosphonates (understood to include bisphosphonates, diphosphonates, bisphosphonic acids and diphosphonic acids).
  • bisphosphonates include but are not limited to: etidronate (Didronel), pamidronate (Aredia), alendronate (Fosamax), risedronate (Actonel), zoledronate (Zometa), ibandronate (Boniva), incadronate or cimadronate, clodronate, EB-1053, minodronate, neridronate, piridronate and tiludronate including any and all pharmaceutically acceptable salts, derivatives, hydrates and mixtures thereof.
  • a compound of the instant invention may also be administered with an agent useful in the treatment of anemia.
  • an anemia treatment agent is, for example, a continuous eythropoiesis receptor activator (such as epoetin alfa).
  • a compound of the instant invention may also be administered with an agent useful in the treatment of neutropenia.
  • a neutropenia treatment agent is, for example, a hematopoietic growth factor which regulates the production and function of neutrophils such as a human granulocyte colony stimulating factor, (G-CSF).
  • G-CSF human granulocyte colony stimulating factor
  • Examples of a G-CSF include filgrastim and PEG-filgrastim.
  • a compound of the instant invention may also be administered with an immunologic - enhancing drug, such as levamisole, isoprinosine and Zadaxin.
  • an immunologic - enhancing drug such as levamisole, isoprinosine and Zadaxin.
  • a compound of the instant invention may also be useful for treating or preventing breast cancer in combination with aromatase inhibitors.
  • aromatase inhibitors include but are not limited to: anastrozole, letrozole and exemestane.
  • a compound of the instant invention may also be useful for treating or preventing cancer in combination with other siNA therapeutics.
  • the compounds of the instant invention may also be administered in combination with ⁇ -secretase inhibitors and/or inhibitors of NOTCH signaling.
  • Such inhibitors include compounds described in WO 01/90084, WO 02/30912, WO 01/70677, WO 03/013506, WO 02/36555, WO 03/093252, WO 03/093264, WO 03/093251, WO 03/093253, WO 2004/039800, WO 2004/039370, WO 2005/030731, WO 2005/014553, USSN 10/957,251, WO 2004/089911, WO 02/081435, WO 02/081433, WO 03/018543, WO 2004/031137, WO 2004/031139, WO 2004/031138, WO 2004/101538, WO 2004/101539 and WO 02/47671 (including LY-450139).
  • a compound of the instant invention may also be useful for treating or preventing cancer in combination with PARP inhibitors.
  • a compound of the instant invention may also be useful for treating cancer in combination with the following therapeutic agents: abarelix (Plenaxis depot®); aldesleukin (Prokine®); Aldesleukin (Proleukin®); Alemtuzumabb (Campath®); alitretinoin (Panretin®); SIR-ONC-00001 allopurinol (Zyloprim®); altretamine (Hexalen®); amifostine (Ethyol®); anastrozole (Arimidex®); arsenic trioxide (Trisenox®); asparaginase (Elspar®); azacitidine (Vidaza®); bendamustine hydrochloride (Treanda®); bevacuzimab (Avastin®); bexarotene capsules (Targretin®); bexarotene gel (Targretin®); bleomycin (Blenoxane®); bortezomib
  • the invention also provides a combination comprising an siNA molecule of the invention comprising at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A) and/or a pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof together with another MET inhibitor.
  • compositions comprising a combination as defined above together with a pharmaceutically acceptable diluent or carrier represent a further aspect of the invention.
  • the individual compounds of such combinations can be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations. In one embodiment, the individual compounds will be administered simultaneously in a combined pharmaceutical formulation.
  • the described molecules could be used in combination with one or more known compounds, treatments, or procedures to prevent or treat diseases, disorders, conditions, and traits described herein in a subject or organism as are known in the art, such as other MET inhibitors.
  • one aspect of the invention comprises a method of treating a subject including, but not limited to, a human suffering from a condition which is mediated by the action, or by loss of action, of MET gene expression, which method comprises administering to said subject an effective amount of a double- stranded siNA molecule of the invention.
  • the siNA molecules comprises at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A).
  • the condition is or is caused by cancer.
  • the molecules and compositions of the instant invention are useful in a method for treating cancer.
  • Cancers treatable according to this aspect of the invention Cardiac: sarcoma fibrosarcoma, rhabdomyosarcoma, and liposarcoma;
  • Lung bronchogenic carcinoma, squamous cell carcinoma, undifferentiated small cell carcinoma, undifferentiated large cell carcinoma, lung adenocarcinoma, alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous harmartoma, mesothelioma, non-small cell lung carcinoma, small cell lung cancer, and lung carcinoma;
  • Gastrointestinal esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma), stomach (carcinoma, leiomyosarcoma), pancreatic cancer (ductal adenocarcinoma, gastrinoma),
  • Liver hepatoma, hepatocellular carcinoma, cholangiocarcinoma, hepatoblastoma, and liver carcinoma
  • Bone osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, multiple myeloma, malignant giant cell tumor benign chondroma, giant cell tumors, osteosarcoma (OS), chordoma, Synovial Sarcoma, and Clear-cell sarcoma
  • Nervous system meninges (meningioma, gliomatosis), brain (astrocytoma, glioblastoma), spinal cord (neurofibroma, meningioma, glioma), neuroblastoma, meningioma, neurofibroma, schwannoma, and esophogeal carcinoma
  • Gynecological sarcoma osteosarcoma
  • the siNA molecules of the instant invention are useful in a method for treating or preventing cancer selected from: head and neck squamous cell carcinomas, histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, non- small cell lung cancer, pancreatic cancer, colorectal cancer, cervical cancer, liver cancers, gastric cancers, colon cancers, glioblastomas, thyroid cancers and breast carcinoma.
  • the compounds of the instant invention are useful in a method for treating or preventing cancer selected from: histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, pancreatic cancer, liver cancers, gastric cancers, colon cancers, multiple myeloma, glioblastomas and breast carcinoma.
  • the compounds of the instant invention are useful for treating liver cancer, such as hepatoma, hepatocellular carcinoma, cholangiocarcinoma, hepatoblastoma, and liver carcinoma.
  • the siNA molecules of the instant invention are useful in a method for the prevention or modulation of the metastases of cancer cells and cancer.
  • the siNA molecules of the instant invention are useful in a method to prevent or modulate the metastases of ovarian cancer, childhood hepatocellular carcinoma, metastatic head and neck squamous cell carcinomas, gastric cancers, breast cancer, colorectal cancer, cervical cancer, lung cancer, nasopharyngeal cancer, pancreatic cancer, glioblastoma and sarcomas.
  • the administration of the siNA molecule is via local administration or systemic administration.
  • the invention features contacting the subject or organism with an siNA molecule of the invention via local administration to relevant tissues or cells, such as lung cells and tissues, such as via pulmonary delivery.
  • the invention features contacting the subject or organism with an siNA molecule of the invention via systemic administration (such as via intravenous or subcutaneous administration of siNA) to relevant tissues or cells, such as cancerous tissues or cells in a subject or organism.
  • siNA molecules of the invention are also used as reagents in ex vivo applications.
  • siNA reagents are introduced into tissue or cells that are transplanted into a subject for therapeutic effect.
  • the cells and/or tissue can be derived from an organism or subject that later receives the explant, or can be derived from another organism or subject prior to transplantation.
  • the siNA molecules can be used to modulate the expression of one or more genes in the cells or tissue, such that the cells or tissue obtain a desired phenotype or are able to perform a function when transplanted in vivo.
  • certain MET target cells from a patient are extracted.
  • These extracted cells are contacted with MET siNAs targeting a specific nucleotide sequence within the cells under conditions suitable for uptake of the siNAs by these cells ⁇ e.g., using delivery reagents such as cationic lipids, liposomes and the like or using techniques such as electroporation to facilitate the delivery of siNAs into cells).
  • delivery reagents such as cationic lipids, liposomes and the like or using techniques such as electroporation to facilitate the delivery of siNAs into cells.
  • the cells are then reintroduced back into the same patient or other patients.
  • a pharmaceutically effective dose of the siNA molecules or pharmaceutical compositions of the invention is administered to the subject.
  • a pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat SIR-ONC-00001
  • a therapeutically effective dose of the siNA of the invention to be administer to a given subject, by taking into account factors, such as the size and weight of the subject, the extent of the disease progression or penetration, the age, health, and sex of the subject, the route of administration m and whether the administration is regional or systemic. Generally, an amount between 0.1 mg/kg and 100 mg/kg body weight/day of active ingredients is administered dependent upon potency of the negatively charged polymer.
  • the siNA molecules of the invention can be administered in a single dose or in multiple doses.
  • siNA molecules of the instant invention can be administered once monthly, once weekly, once daily (QD), or divided into multiple monthly, weekly, or daily doses, such as, for example, but not limitation, twice daily (BID), three times daily (TID), once every two weeks.
  • QD once weekly, once daily
  • TID three times daily
  • the administration can be continuous, i.e., every day, or intermittently.
  • intermittent administration of a compound of the instant invention may be administration one to six days per week or it may mean administration in cycles (e.g. daily administration for two to eight consecutive weeks, then a rest period with no administration for up to one week) or it may mean administration on alternate days.
  • compositions or formulations can be administered in a variety of ways.
  • administration methods of the invention include oral, buccal, sublingual, parenteral (i.e., intraarticularly, intravenously, intraperitoneally, subcutaneously, or intramuscularly), local rectal administration or other local administration.
  • the composition of the invention can be administered by insufflation and inhalation. Administration can be accomplished via single or divided doses.
  • the pharmaceutical compositions are administered intravenously or intraperitoneally by a bolus injection (see, e.g., U.S. Pat. No. 5,286,634).
  • Lipid nucleic acid particles can be administered by direct injection at the site of disease or by injection at a site distal from the site of disease (see, e.g., Culver, HUMAN GENE THERAPY, MaryAnn Liebert, Inc., Publishers, New York. pp. 70-71(1994)).
  • the siNA molecules of the invention and formulations or compositions SIR-ONC-00001 thereof are administered to a cell, subject, or organism as is described herein and as is generally known in the art.
  • the siNA can be administered to the subject systemically as described herein or otherwise known in the art, either alone as a monotherapy or in combination with additional therapies described herein or as are known in the art.
  • Systemic administration can include, for example, pulmonary (inhalation, nebulization etc.) intravenous, subcutaneous, intramuscular, catheterization, nasopharangeal, transdermal, or oral/gastrointestinal administration as is generally known in the art.
  • the siNA can be administered to the subject locally or to local tissues as described herein or otherwise known in the art, either alone as a monotherapy or in combination with additional therapies as are known in the art.
  • Local administration can include, for example, inhalation, nebulization, catheterization, implantation, direct injection, dermal/transdermal application, patches, stenting, ear/eye drops, or portal vein administration to relevant tissues, or any other local administration technique, method or procedure, as is generally known in the art.
  • the siNA molecules of the invention and formulations or compositions thereof are administered to the liver as is generally known in the art (see for example Wen et al, 2004, World J Gastroenterol., 10, 244-9; Murao et ah, 2002, Pharm Res., 19, 1808-14; Liu et al, 2003, gene Ther., 10, 180-7; Hong et al, 2003, / Pharm Pharmacol, 54, 51-8; Herrmann et al., 2004, Arch Virol., 149, 1611-7; and Matsuno et al., 2003, gene Ther., 10, 1559-66).
  • the invention features the use of methods to deliver the siNA molecules of the instant invention to hematopoietic cells, including monocytes and lymphocytes. These methods are described in detail by Hartmann et al., 1998, J. Phamacol. Exp. Ther., 285(2), 920-928; Kronenwett et al, 1998, Blood, 91(3), 852-862; Filion and Phillips, 1997, Biochim. Biophys. Acta., 1329(2), 345-356; Ma and Wei, 1996, Leuk. Res., 20(11/12), 925-930; and Bongartz et al, 1994, Nucleic Acids Research, 22(22), 4681-8. SIR-ONC-00001
  • the siNA molecules of the invention and formulations or compositions thereof are administered directly or topically (e.g., locally) to the dermis or follicles as is generally known in the art (see for example Brand, 2001, Curr. Opin. Mol. Ther., 3, 244-8; Regnier et al., 1998, J. Drug Target, 5, 275-89; Kanikkannan, 2002, BioDrugs, 16, 339-47; Wraight et al, 2001, Pharmacol. Ther., 90, 89-104; and Preat and Dujardin, 2001, STP PharmaSciences, 11, 57-68).
  • the siNA molecules of the invention and formulations or compositions thereof are administered directly or topically using a hydroalcoholic gel formulation comprising an alcohol (e.g., ethanol or isopropanol), water, and optionally including additional agents such isopropyl myristate and carbomer 980.
  • a hydroalcoholic gel formulation comprising an alcohol (e.g., ethanol or isopropanol), water, and optionally including additional agents such isopropyl myristate and carbomer 980.
  • the siNA are formulated to be administered topically to the nasal cavity.
  • Topical preparations can be administered by one or more applications per day to the affected area; over skin areas occlusive dressings can advantageously be used. Continuous or prolonged delivery can be achieved by an adhesive reservoir system.
  • an siNA molecule of the invention is administered iontophoretically, for example to a particular organ or compartment (e.g., the eye, back of the eye, heart, liver, kidney, bladder, prostate, tumor, CNS etc.).
  • a particular organ or compartment e.g., the eye, back of the eye, heart, liver, kidney, bladder, prostate, tumor, CNS etc.
  • iontophoretic delivery are described in, for example, WO 03/043689 and WO 03/030989, which are incorporated by reference in their entireties herein.
  • the siNA molecules of the invention and formulations or compositions thereof are administered to the lung as is described herein and as is generally known in the art.
  • the siNA molecules of the invention and formulations or compositions thereof are administered to lung tissues and cells as is described in U.S. Patent Publication Nos. 2006/0062758; 2006/0014289; and 2004/0077540.
  • Aerosols and Delivery Devices a. Aerosol Formulations
  • compositions of the present invention can be made into aerosol formulations (i.e., they can be "nebulized") to be administered via inhalation (e.g., intranasally or intratracheally) (see, Brigham et al., Am. J. Set, SIR-ONC-00001
  • Aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • the siNA molecules of the invention and formulations thereof are administered via pulmonary delivery, such as by inhalation of an aerosol or spray dried formulation administered by an inhalation device or nebulizer, providing rapid local uptake of the nucleic acid molecules into relevant pulmonary tissues.
  • Solid particulate compositions containing respirable dry particles of micronized nucleic acid compositions can be prepared by grinding dried or lyophilized nucleic acid compositions, and then passing the micronized composition through, for example, a 400 mesh screen to break up or separate out large agglomerates.
  • a solid particulate composition comprising the siNA compositions of the invention can optionally contain a dispersant which serves to facilitate the formation of an aerosol as well as other therapeutic compounds.
  • a suitable dispersant is lactose, which can be blended with the nucleic acid compound in any suitable ratio, such as a 1 to 1 ratio by weight.
  • Spray compositions comprising siNA molecules or compositions of the invention can, for example, be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurized packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant.
  • aerosol compositions of the invention suitable for inhalation can be either a suspension or a solution and generally contain an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A), and a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1, 2,3,3, 3-heptafluoro-n-propane or a mixture thereof.
  • a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1, 2,3,3, 3-heptaflu
  • the aerosol composition can optionally contain additional formulation excipients well known in the art such as surfactants.
  • additional formulation excipients well known in the art such as surfactants.
  • Non-limiting examples include oleic acid, lecithin or an oligolactic acid or derivative such as those described in W094/21229 and W098/34596 and co-solvents for example ethanol.
  • a pharmaceutical aerosol formulation of the invention comprising a compound of the invention and a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof as propellant, optionally in combination with a surfactant and/or a co- solvent.
  • aerosol formulations of the invention can be buffered by the addition of suitable buffering agents.
  • Aerosol formulations can include optional additives including preservatives if the formulation is not prepared sterile.
  • preservatives include, methyl hydroxybenzoate, anti-oxidants, flavorings, volatile oils, buffering agents and emulsifiers and other formulation surfactants.
  • fluorocarbon or perfluorocarbon carriers are used to reduce degradation and provide safer biocompatible non-liquid particulate suspension compositions of the invention (e.g., siNA and/or LNP formulations thereof).
  • a device comprising a nebulizer delivers a composition of the invention (e.g., siNA and/or LNP formulations thereof) comprising fluorochemicals that are bacteriostatic thereby decreasing the potential for microbial growth in compatible devices.
  • a composition of the invention e.g., siNA and/or LNP formulations thereof
  • Capsules and cartridges comprising the composition of the invention for use in an inhaler or insufflator, of for example gelatine, can be formulated containing a powder mix for inhalation of a compound of the invention and a suitable powder base such as lactose or starch.
  • each capsule or cartridge contains an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101 ; or formula (A), and one or more excipients.
  • the compound of the invention can be presented without excipients such as lactose
  • the aerosol compositions of the present invention can be administered into the respiratory system as a formulation including particles of respirable size, e.g. particles of a size sufficiently small to pass through the nose, mouth and larynx upon inhalation and through the bronchi and alveoli of the lungs.
  • respirable particles range from about 0.5 to 10 microns in size.
  • the particulate range can be from 1 to 5 microns.
  • the particulate range can be from 2 to 3 microns. Particles of non- respirable size which are included in the aerosol tend to deposit in the throat and be swallowed, and the quantity of non-respirable particles in the aerosol is thus minimized.
  • a particle size in the range of 10-500 um is preferred to ensure retention in the nasal cavity.
  • an siNA composition of the invention is administered topically to the nose for example, for the treatment of rhinitis, via pressurized aerosol formulations, aqueous formulations administered to the nose by pressurized pump or by nebulization.
  • Suitable formulations contain water as the diluent or carrier for this purpose.
  • the aqueous formulations for administration of the composition of the invention to the lung or nose can be provided with conventional excipients such as buffering agents, tonicity modifying agents and the like.
  • siNA molecules of the invention can be formulated and delivered as particles and/or aerosols as discussed above and dispensed from various aerosolization devices known by those of skill in the art.
  • Aerosols of liquid or non-liquid particles comprising an siNA molecule or formulation of the invention can be produced by any suitable means, such as with a device comprising a nebulizer (see for example US 4,501,729) such as ultrasonic or air jet nebulizers.
  • a nebulizer see for example US 4,501,729
  • ultrasonic or air jet nebulizers such as ultrasonic or air jet nebulizers.
  • Solid particle aerosols comprising an siNA molecule or formulation of the invention and surfactant can be produced with any solid particulate aerosol generator.
  • One type of solid particle aerosol generator used with the siNA molecules of the invention is an insufflator.
  • a second type of illustrative aerosol generator comprises a metered dose inhaler ("MDI"). MDIs containing siNA molecules or formulations taught herein can be prepared by methods of the art (for example, see Byron, above and WO96/32099).
  • siNA molecules can also be formulated as a fluid formulation for delivery from a fluid dispenser, such as those described and illustrated in WO05/044354.
  • nebulizer devices are used in applications for conscious, spontaneously breathing subjects, and for controlled ventilated subjects of all ages.
  • the nebulizer devices can be used for targeted topical and systemic drug delivery to the lung.
  • a device comprising a nebulizer is used to deliver an siNA molecule or formulation of the invention locally to lung or pulmonary tissues.
  • a SIR-ONC-00001 device comprising a nebulizer is used to deliver a an siNA molecule or formulation of the invention systemically.
  • siNA molecules of the invention can also be used for diagnostic applications, research applications, and/or manufacture of medicants.
  • the invention features a method for diagnosing a disease, trait, or condition in a subject comprising administering to the subject a composition of the invention under conditions suitable for the diagnosis of the disease, trait, or condition in the subject.
  • siNA molecules of the invention are used to down regulate or inhibit the expression of MET proteins arising from haplotype polymorphisms that are associated with a trait, disease or condition in a subject or organism.
  • Analysis of MET genes, or MET protein or RNA levels can be used to identify subjects with such polymorphisms or those subjects who are at risk of developing traits, conditions, or diseases described herein. These subjects are amenable to treatment, for example, treatment with siNA molecules of the invention and any other composition useful in treating diseases related to target gene expression.
  • analysis of MET protein or RNA levels can be used to determine treatment type and the course of therapy in treating a subject.
  • Monitoring of MET protein or RNA levels can be used to predict treatment outcome and to determine the efficacy of compounds and compositions that modulate the level and/or activity of certain MET proteins associated with a trait, disorder, condition, or disease.
  • the invention comprises use of a double-stranded nucleic acid according to the invention for use in the manufacture of a medicament.
  • the medicament is for use in treating a condition that is mediated by the action, or by loss of action, of MET.
  • the medicament is for use for the treatment of cancer.
  • the medicament is for use for the treatment of head and neck squamous cell carcinomas, histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, non- small cell lung cancer, pancreatic cancer, papillary renal cell carcinoma, liver cancer, gastric cancer, colon cancer, multiple myeloma, glioblastomas and breast carcinoma.
  • the SIR-ONC-00001 use is for the treatment of liver cancer selected from the group consisting hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma, carcinoma.
  • liver cancer selected from the group consisting hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma, carcinoma.
  • siNAs wherein at least one strand comprises at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A), are for use in a method for treating a cancer, such as, for example but not limitation, of head and neck squamous cell carcinomas, histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, non-small cell lung cancer, pancreatic cancer, papillary renal cell carcinoma, liver cancer, gastric cancer, colon cancer, multiple myeloma, glioblastomas and breast carcinoma.
  • a cancer such as, for example but not limitation, of head and neck squamous cell carcinomas, histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, non-small
  • siNA molecules were designed, synthesized and evaluated for efficacy against MET gene expression. Certain MET sequences were designed and selected by methods set forth in U.S. Application No. 60/182,605. Other sequences were designed and selected using a proprietary algorithm. The primary criteria for design of certain of the MET sequences for human siNAs were (i) homology between two species (human and rhesus monkey) and (ii) high efficacy scores as determined by a proprietary algorithm. The effects of the siNAs on MET RNA levels.
  • the target sequences of the siNAs that were selected are set forth in Table la (target sequences).
  • the sense and antisense strands of the siNA sequences corresponding to the target sequences in Table la are set forth in Table lb.
  • Various chemically modified siNAs that were synthesized are set forth in Table lc.
  • Table la MET Target Sequences, noting target sites. SIR-ONC-00001
  • the two individual, complementary strands of the siNA were synthesized separately using solid phase synthesis, then purified separately by reversed phase solid phase extraction (SPE).
  • SPE reversed phase solid phase extraction
  • the complementary strands were annealed to form the double strand (duplex) and delivered in the desired concentration and buffer of choice.
  • the single strand oligonucleotides were synthesized using phosphoramidite chemistry on an automated solid-phase synthesizer, using procedures as are generally known in the art (see for example U.S. Application No. 12/064,014).
  • a synthesis column was packed with solid support derivatized with the first nucleoside residue (natural or chemically modified). Synthesis was initiated by detritylation of the acid labile 5'-0-dimethoxytrityl group to release the 5'-hydroxyl.
  • a suitably protected phosphoramidite and a suitable activator in acetonitrile were delivered simultaneously to the synthesis column resulting in coupling of the amidite to the SIR-ONC-00001
  • the trityl-on solution of each crude single strand was purified using chromatographic purification, such as SPE RPC purification.
  • the hydrophobic nature of the trityl group permits stronger retention of the desired full-length oligo than the non-tritylated truncated failure sequences.
  • the failure sequences were selectively washed from the resin with a suitable solvent, SIR-ONC-00001 such as low percent acetonitrile. Retained oligonucleotides were then detritylated on-column with trifluoroacetic acid to remove the acid-labile trityl group.
  • Annealing Based on the analysis of the product, the dried oligos were dissolved in appropriate buffers followed by mixing equal molar amounts (calculated using the theoretical extinction coefficient) of the sense and antisense oligonucleotide strands. The solution was then analyzed for purity of duplex by chromatographic methods and desired final concentration. If the analysis indicated an excess of either strand, then the additional non-excess strand was titrated until duplexing was complete. When analysis indicated that the target product purity has been achieved the material was delivered and ready for use.
  • A, C, G, and U ribose A, C, G or U
  • Ultrafiltration The annealed product solution is concentrated using a TFF system containing an appropriate molecular weight cut-off membrane. Following concentration, the product solution is desalted via diafiltration using Milli-Q water until the conductivity of the filtrate is that of water.
  • Lyophilization The concentrated solution is transferred to a bottle, flash frozen and attached to a lyophilizer. The product is then freeze-dried to a powder. The bottle is removed from the lyophilizer and is now ready for use.
  • Human hepatoma cell line, HepG2, rhesus kidney epithelial cell line, LLC-MK2 Derivative, and cynomulgus skin fibroblast cell line, Cynom-Kl were grown in modified Eagle's SIR-ONC-00001 medium. All the culture media were supplemented with 10% fetal bovine serum, 100 ⁇ g/mL streptomycin, lOOU/mL penicillin, and 1% sodium bicarbonate.
  • Cells were plated in all wells of tissue-culture treated, 96-well plates at a final count of 3500 (HepG2 and LLC-MK2 Derivative) or 5000 (Cynom-Kl) cells/well in ⁇ -. of the appropriate culture media. For duration assays, HepG2 cells were plated at 1200 cell/swell in ⁇ medium. The cells were cultured for overnight after plating at 37°C in the presence of 5% C0 2 .
  • RNAiMax complexes containing siNA and RNAiMax (Invitrogen) were created as follows. A solution of RNAiMax diluted 33-fold in OPTI-MEM was prepared. In parallel, solutions of the siNAs for testing were prepared to a final concentration of 120 nM in OPTI-MEM. After incubation of RNAiMax/OPTI-MEM solution at room temperature for 5 min, an equal volume of the siNA solution and the RNAiMax solution were added together for each of the siNAs.
  • siNAs were transfected at 200, 150, 100 or 75pM per well.
  • siNA series are 6-fold serial dilution starting at 30nM or 4-fold serial dilution starting at 40nM. All transfections were set up as multiple biological replicates.
  • RNAiMax -siNA complexes The time of incubation with the RNAiMax -siNA complexes was 24 hours and there was no change in media between transfection and harvesting for screening and dose response curve studies. For duration assays, the time of incubation with the RNAiMax -siNA complexes was 24, 72, and 120 hours. There was no change in media between transfection and harvesting for 24- and 72-hour time points. Media was replaced with fresh media 72 hours after transfection for 120-hour time point.. SIR-ONC-00001
  • the culture medium was aspirated and discarded from the wells of the culture plates at the desired time points.
  • the transfected cells were washed once with 50uL DPBS solution per well.
  • Fifty microliters per well of the Lysis Solution from the TaqMan® Gene Expression Cells- to-CTTM Kit (Applied Biosystems, Cat# 4399002) supplemented with DNase I was added directly to the plates to lyse the cells.
  • Five microliters per well of Stop Solution from the same kit was added to the plates to inactivate DNase I 5 min. later.
  • the lysis plates were incubated for at least 2 min. at room temperature. The plates can be stored for 2 hours at 4°C, or -80 °C for two months.
  • Each well of the reverse transcription plate required lOuL of 2X reverse transcriptase buffer, luL of 20X reverse transcription enzyme and 2uL of nuclease-free water.
  • the reverse transcription master mix was prepared by mixing 2X reverse transcription buffer, 20X reverse transcription enzyme mix, and nuclease-free water. 13uL of the reverse transcription master mix was dispensed into each well of the reverse transcription plate (semi-skirted). A separate reverse transcription plate was prepared for each cell plate. A separate reverse transcription plate was prepared for each cell plate. Seven microliters per lysate from the cell lysis procedure described above was added into each well of the reverse transcription plate.
  • the plate was sealed and spun on a centrifuge (lOOOrpm for 30 seconds) to settle the contents to the bottom of the reverse transcription plate.
  • the plate was placed in a thermocycler at 37 °C for 60 min, 95 °C for 5 min, and 4 °C until the plate is removed from the thermocycler. Upon removal, if not used immediately, the plate was frozen at -20 °C.
  • Table 2 Probes and primers used to carry out Real-Time RT/PCR (Taqman) reactions for MET mRNA analysis.
  • the assays were performed on an ABI 7900 instrument, according to the manufacturer's instructions.
  • a TaqMan Gene Expression Master Mix (provided in the Cells-to- CTTM Kit, Applied Biosystems, Cat # 4399002) was used.
  • the PCR reactions were carried out at 50 °C for 2 min, 95 °C for 10 min followed by 40 cycles at 95 °C for 15 sees and 60 °C for 1 min.
  • the baseline was set in the exponential phase of the amplification curve, and based on the intersection point of the baselines with the amplification curve, a Ct value was assigned by the instrument.
  • AACt (log2(fold change)) ACt (Taiget siNA ) - ACt (NT c)
  • the non-targeting control siNA was, unless otherwise indicated, chosen as the value against which to calculate the percent inhibition (knockdown) of gene expression, because it is the most relevant control.
  • the level of protein was quantified using the Bio-Rad VersaDoc Imager according to the protocols of that piece of equipment. A pixel count was performed in each lane using an area of identical size. Each sample was then compared to the appropriate control treated sample and converted to a percent of protein remaining compared to control.
  • the MET siNAs were designed and synthesized as described previously. Various siNAs were screened in HepG2 cells. The log 2(fold change) in MET gene expression data upon treatment with various modified MET siNAs in human cells is shown in Table 3a and b. Each screen was performed at 24 hrs. Quantitative RT-PCR was used to assess the level of MET mRNA and the data were normalized to the expression level of GAPDH (an ubiquitously expressed 'house-keeping' gene). Each treatment was then normalized against the non-MET targeting control.
  • GAPDH an ubiquitously expressed 'house-keeping' gene
  • siNAs from Table 3a and Table 3b having a large log2(fold change) in the primary screen and/or a low IC 50 in dose response curves were tested in human HepG2 cells at a lower concentrations to determine the effect of the MET siNAs on the mRNA transcript of SIR-ONC-00001 human MET at varying concentrations.
  • Table 4 summarizes the data. The siNAs were ranked based on log2(fold change) in MET mRNA expression level.
  • siNAs from Table 4 were further analyzed for efficacy and potency in HepG2cells, LLC-MK2 Derivative and Cynom-Kl cells to ensure that these siNAs functioned well in monkey cells for primate studies.
  • the results for SIR-ONC-00001 these siNAs are shown in Table 5.
  • the potency 50 is the calculated siNA transfection concentration that produces 50% target mRNA knockdown.
  • Table 5 Dose response data for various siNAs in HepG2 -Human hepatoma, LLC-MK2 Derivative - Rhesus monkey kidney epithelial, and Cynom-Kl - Cynomolgus monkey skin fibroblast cell lines. Calculated maximum AACt is determined from the dose response curve.
  • siNAs were reconstituted as 50 ⁇ to ⁇ stock solution with H 2 0 and added to human serum pre- warmed to 37 °C to a final concentration of 20 ⁇ g/mL. The mixture was then incubated at 37°C for 0, 1 and 2 hours. At the end of each time point, the reactions were stopped by mixing with equal volume of Phenomenex Lysis-Loading Buffer. Oligonucleotides were purified in 96-well format by Phenomenex Solid Phase Extraction and lyophilized until dry with Labconco Triad Lyo-00417. The lyophilized samples were reconstituted in 150 ⁇ ⁇ of 1 mM EDTA prepared with RNase-free H 2 0.
  • sample solutions were then diluted 5 fold with 1 mM EDTA for liquid chromatography/mass spectrometry (LC/MS) analysis on Thermo Fisher Orbitrap. Serum metabolites of the siNAs were determined based on the measured molecular weights.
  • LC/MS liquid chromatography/mass spectrometry
  • siNAs targeting these target sites were synthesized with a series of additional modification motifs and tested in a primary screen to SIR-ONC-00001 determine knockdown. Select compounds from the primary screen (those demonstrating a large log2(fold change)) were then tested in low concentration screens and/or in a human serum stability assays.
  • siNAs tested in the human serum stability assays were tested to determine the percent of the full-length sense and antisense siNA strands remaining after the time periods of 1 hour and 2 hours.
  • the log2(fold change) data for these siNAs are shown in Table 6.
  • the four compounds that had the best overall knockdown (i.e., largest log2(fold change) from the primary and low concentration assays combined were selected along with a control siNA having Stab7/Stab35G chemistry for testing with regard to human serum stability at 1 and 2 hour intervals.
  • the cytokines and chemokines measured are IL-la, IL- ⁇ , IL-6, KC, IL-10, IFNy, TNF, GMCSF, ⁇ - ⁇ , MCP-l/JE, and RANTES.
  • the levels of inflammatory cytokines (exp. KC, IFN-g, TNF-a, IL-6) were not induced above the levels of delivery vehicle itself for the siNAs tested, specifically R-
  • mice were dosed IV via tail vein injections with LNP encapsulated siNAs or vehicle control using 2 different 3-week dosing schemes: one 1 mg/kg dose for 3 consecutive days or a single 6 mg/kg does per week.
  • the mice were co-dosed with sorafenib at a dose of 100 mg/kg BID every day for 3 weeks.
  • Total tumor burden was measured by micro-CT scan imaging. The animals were sacrificed 5 days after the last siNA dose (Day 23), and normal liver and tumor tissues from each animal were collected for RNA purification. Total RNA was purified using RNeasy 96 kit (Qiagen, Cat# 74182).
  • cDNA was generated from total RNA using High Capacity cDNA Reverse Transcription Kit (Cat#: 4368813). Quantitative PCR reactions were performed with TaqMan Universal PCR Master Mix (Cat#: 4304437). Human MET TaqMan Gene Expression Assay (Hs00179845_ml) and mouse GAPDH TaqMan Gene Expression Assay (Cat#: 4352932E) were used to monitor the mRNA level of both transcripts. The expression level of MET was normalized against GAPDH to minimize technical variations. All reverse transcription and quantitative PCR reagents are from Applied Biosystems, Inc.
  • a non-human primate pharmacodynamic study was performed in Rhesus macaque monkeys at New Iberia Research Center of University of Louisiana at Lafayette. The animals were dosed with a single 2.5mpk dose of siNA loaded lipid nanoparticles through intravenous infusion.
  • liver biopsies were performed at various time points pre- and post-dose with 16T gauge Menghini needles for about 20mg tissue per animal.
  • Whole blood and serum/plasma were also collected at different time points pre- and post-dose to monitor potential toxicity associated with the treatments. All procedures adhere to the regulations outlined in the USDA Animal Welfare Act (9 CFR, Parts 1, 2 and 3) and the conditions specified in The Guide for Care and Use of Laboratory Animals (ILAR publication, 1996, National Academy Press).
  • LNP formulations (DLinDMA/Cholesterol/S-PEG-C-DMA/DSPC in a 40/48/2/10 ratio) comprising several modified siNAs R-008347048-000B, R-008313708-000E, and R- 008313730-000M in an N/P ratio of 6 were tested.
  • Log2(fold change) in MET gene expression was determined on days 3, 7, 14, and 28 days post-dosing.
  • Pre-dose MET expression levels for the monkeys were measured 7 days before the first dosing. As can be seen from Figure 12, the log 2(fold change) is greater than 4 and the percent inhibition of MET gene expression by these siNAs is greater than 90% at day 3. At day 14, this level of greater than 90% inhibition is still maintained by siNA R-008313708-000E.
  • the lipid nanoparticles were prepared by an impinging jet process.
  • the particles were formed by mixing lipids dissolved in alcohol with siNA dissolved in a citrate buffer.
  • the mixing ratio of lipids to siNA is targeted at 45-55% lipid and 65-45% siNA.
  • the lipid solution SIR-ONC-00001 contained a cationic lipid, a helper lipid (cholesterol), PEG (e.g. PEG-C-DMA, PEG-DMG) lipid, and DSPC at a concentration of 5-15 mg/mL with a target of 9-12 mg/mL in an alcohol (for example ethanol).
  • the ratio of the lipids had a mole percent range of 25-98 for the cationic lipid with a target of 35-65, the helper lipid had a mole percent range from 0-75 with a target of 30-50, the PEG lipid has a mole percent range from 1-15 with a target of 1-6, and the DSPC had a mole percent range of 0-15 with a target of 0-12.
  • the siNA solution contained one or more siNA sequences at a concentration range from 0.3 to 1 .0 mg/mL with a target of 0.3 -0.9 mg/mL in a sodium citrate buffered salt solution with pH in the range of 3.5-5.
  • the two liquids were heated to a temperature in the range of 15-40°C, targeting 30-40°C, and then mixed in an impinging jet mixer instantly forming the LNP.
  • the teelD had a range from 0.25 to 1.0 mm and a total flow rate from 10 -600 mL/min.
  • the combination of flow rate and tubing ID had the effect of controlling the particle size of the LNPs between 30 and 200 nm.
  • the solution was then mixed with a buffered solution at a higher pH with a mixing ratio in the range of 1: 1 to 1:3 vol: vol but targeting 1:2 vol: vol. This buffered solution was at a temperature in the range of 15- 40°C, targeting 30-40°C.
  • the mixed LNPs were held from 30 minutes to 2 hrs prior to an anion exchange filtration step.
  • the temperature during incubating was in the range of 15-40°C, targeting 30-40°C.
  • the solution was filtered through a 0.8 um filter containing an anion exchange separation step.
  • This process used tubing IDs ranging from 1 mm ID to 5 mm ID and a flow rate from 10 to 2000 mL/min.
  • the LNPs were concentrated and diafiltered via an ultrafiltration process where the alcohol was removed and the citrate buffer was exchanged for the final buffer solution such as phosphate buffered saline.
  • the ultrafiltration process used a tangential flow filtration format (TFF). This process used a membrane nominal molecular weight cutoff range from 30 -500 KD.
  • the membrane format was hollow fiber or flat sheet cassette.
  • the TFF processes with the proper molecular weight cutoff retained the LNP in the retentate and the filtrate or permeate contained the alcohol; citrate buffer; and final buffer wastes.
  • the TFF process is a multiple step process with an initial concentration to a siNA concentration of 1 -3 mg/mL. Following concentration, the LNPs solution was diafiltered against the final buffer for 10 -20 volumes to remove the alcohol and perform buffer exchange. The material was then concentrated an additional 1-3 fold. The final steps of the LNP process were to sterile filter the concentrated LNP solution and vial the product.
  • siNA duplex concentrations were determined by Strong Anion-Exchange High- Performance Liquid Chromatography (SAX-HPLC) using Waters 2695 Alliance system (Water Corporation, Milford MA) with a 2996 PDA detector.
  • the LNPs otherwise referred to as RNAi Delivery Vehicles (RDVs)
  • RDVs RNAi Delivery Vehicles
  • Mobile phase was composed of A: 25 mM NaC10 4 , 10 mM Tris, 20% EtOH, pH 7.0 and B: 250 mM NaC10 4 , 10 mM Tris, 20% EtOH, pH 7.0 with a liner gradient from 0-15 min and a flow rate of 1 ml/min.
  • the siNA amount was determined by comparing to the siNA standard curve.
  • Fluorescence reagent SYBR Gold was employed for RNA quantitation to monitor the encapsulation rate of RDVs.
  • RDVs with or without Triton X-100 were used to determine the free siNA and total siNA amount.
  • the assay is performed using a SpectraMax M5e microplate spectrophotometer from Molecular Devices (Sunnyvale, CA). Samples were excited at 485 nm and fluorescence emission was measured at 530 nm. The siNA amount is determined by comparing to an siNA standard curve.
  • Encapsulation rate (1- free siN A/total siNA) xl00%
  • RDVs containing 1 ⁇ g siNA were diluted to a final volume of 3 ml with 1 x PBS.
  • the particle size and polydispersity of the samples was measured by a dynamic light scattering method using ZetaPALS instrument (Brookhaven Instruments Corporation, Holtsville, NY).
  • the scattered intensity was measured with He-Ne laser at 25°C with a scattering angle of 90°.
  • RDVs containing 1 ⁇ g siNA were diluted to a final volume of 2 ml with 1 mM Tris buffer (pH 7.4). Electrophoretic mobility of samples was determined using ZetaPALS SIR-ONC-00001 instrument (Brookhaven Instruments Corporation, Holtsville, NY) with electrode and He-Ne laser as a light source. The Smoluchowski limit was assumed in the calculation of zeta potentials.
  • RP-HPLC Performance Liquid Chromatography
  • CAD Corona charged aerosol detector
  • Individual lipids in RDVs were analyzed using an Agilent Zorbax SB- CIS (50 x 4.6 mm, 1.8 ⁇ particle size) column with CAD at 60°C.
  • the mobile phase was composed of A: 0.1% TFA in H 2 0 and B: 0.1% TFA in IPA.
  • the individual lipid concentration was determined by comparing to the standard curve with all the lipid components in the RDVs with a quadratic curve fit. The molar percentage of each lipid was calculated based on its molecular weight.
  • siNA solutions were prepared by dissolving siNAs in 25mM citrate buffer (pH 4.0) at a concentration of 0.8mg/mL.
  • Lipid solutions were prepared by dissolving a mixture of 2S- Octyl-ClinDMA, cholesterol and PEG-DMG at a ratio of 71.9:20.2:7.9 in absolute ethanol at a concentration of about lOmg/mL.
  • Equal volume of siNA and lipid solutions were delivered with two syringe pumps at the same flow rates to a mixing T connector. The resulting milky mixture was collected in a sterile bottle.
  • This mixture was then diluted slowly with an equal volume of citrate buffer, and filtered through a size exclusion hollow fiber cartridge to remove any free siNA in the mixture.
  • Ultra filtration against citrate buffer pH 4.0 was employed to remove SIR-ONC-00001 ethanol (test stick from ALCO screen), and against PBS (pH 7.4) to exchange buffer.
  • the final LNP was obtained by concentrating to a desired volume and sterile filtered through a 0.2mm filter.
  • the obtained LNPs were characterized in term of particle size, alcohol content, total lipid content, nucleic acid encapsulated, and total nucleic acid concentration.
  • siNA nanoparticle solutions in Table 13 are prepared by dissolving siNAs and/or carrier molecules in 25 mM citrate buffer (pH 4.0) at a concentration of 0.9 mg/mL.
  • Lipid solutions are prepared by dissolving a mixture of cationic lipid (e.g., CLinDMA or DOBMA, see structures and ratios for Formulations in Table 13), DSPC, Cholesterol, and PEG-DMG (ratios shown in Table 13) in absolute ethanol at a concentration of about 15 mg/mL.
  • the nitrogen to phosphate ratio is approximate to 3: 1.
  • Equal volume of siNA/carrier and lipid solutions are delivered with two FPLC pumps at the same flow rates to a mixing T connector.
  • a back pressure valve is used to adjust to the desired particle size.
  • the resulting milky mixture is collected in a sterile glass bottle.
  • This mixture is then diluted slowly with an equal volume of citrate buffer, and filtered through an ion- exchange membrane to remove any free siNA/carrier in the mixture.
  • Ultra filtration against citrate buffer (pH 4.0) is employed to remove ethanol (test stick from ALCO screen), and against PBS (pH 7.4) to exchange buffer.
  • the final LNP is obtained by concentrating to a desired volume and sterile filtered through a 0.2 ⁇ filter.
  • the obtained LNPs are characterized in term of particle size, Zeta potential, alcohol content, total lipid content, nucleic acid encapsulated, and total nucleic acid concentration.
  • a LNP-086 siNA/carrier formulation is prepared in bulk as follows. The process consists of (1) preparing a lipid solution; (2) preparing an siNA/carrier solution; (3) mixing/particle formation; (4) incubation; (5) dilution; (6) ultrafiltration and concentration.
  • a 3-necked 2L round bottom flask, a condenser, measuring cylinders, and two 10L conical glass vessels are depyrogenated.
  • the lipids are warmed to room temperature.
  • 50.44g of CLinDMA with a pipette and 43.32g of DSPC 5.32g of Cholesterol , 6.96g of PEG-DMG, and 2.64g of linoleyl alcohol are added.
  • To the mixture is added 1L of ethanol.
  • the round bottom flask is placed in a heating mantle that is connected to a J-CHEM process controller.
  • the lipid suspension is stirred under Argon with a stir bar and a condenser on top.
  • a thermocouple probe is put into the suspension through one neck of the round bottom flask with a sealed adapter.
  • the suspension is heated at 30 °C until it became clear.
  • the solution is allowed to cool to room temperature and transferred to a conical glass vessel and sealed with a cap.
  • a sterile container such as the Corning storage bottle, is weighed 3.6 g times the water correction factor (approximately 1.2) of siNA-1 powder.
  • the siNA is transferred to a depyrogenated 5 L glass vessel.
  • the weighing container is rinsed 3x with citrate buffer (25mM, pH 4.0, and lOOmM NaCl) and the rinses are placed into the 5 L vessel, QS with citrate buffer to 4 L.
  • the concentration of the siNA solution is determined with a UV spectrometer using the following procedure. 20 ⁇ ⁇ is removed from the solution, diluted 50 times to 1000 ⁇ , and the UV reading recorded at A260 nm after blanking with citrate buffer. This is repeated.
  • the concentration is calculated based on the extinction coefficients of the siNAs. If the final concentration are out of the range of 0.90 + 0.01 mg/mL, the concentration is adjusted by adding more siNA/carrier powder, or adding more citrate buffer. This process is repeated for the second siNA, siNA-2.. Into a depyrogenated 10L glass vessel, 4 L of each 0.9 mg/mL siNA solution is transferred.
  • the siNA/carrier solution comprised a single siNA duplex and or carrier instead of a cocktail of two or more siNA duplexes and/or carriers
  • the siNA/carrier is dissolved in 25 mM citrate buffer (pH 4.0, 100 mM of NaCl) to give a final concentration of 0.9 mg/mL.
  • lipid/ethanol solution is then sterile/filtered through a Pall Acropak 20 0.8/0.2 ⁇ sterile filter PN 12203 into a depyrogenated glass vessel using a Master Flex Peristaltic SIR-ONC-00001
  • the filtration process is run at an 80 mL scale with a membrane area of 20 cm .
  • the flow rate is 280 mL/min. This process is scaleable by increasing the tubing diameter and the filtration area.
  • An AKTA P900 pump is turned on and sanitized by placing 1000 mL of 1 N NaOH into a 1 L glass vessel and 1000 mL of 70% ethanol into a 1 L glass vessel and attaching the pump with a pressure lid to each vessel.
  • a 2000 mL glass vessel is placed below the pump outlet and the flow rate is set to 40 mL/min for a 40 minute time period with argon flushing the system at 10 psi.
  • the gas is turned off and the pump is stored in the solutions until ready for use. Prior to use, the pump flow is verified by using 200 mL of ethanol and 200 mL of sterile citrate buffer.
  • AKTA pump To the AKTA pump is attached the sterile lipid/ethanol solution, the sterile siNA/carrier or siNA/carrier cocktail /citrate buffer solution and a depyrogenated receiving vessel (2x batch size) with lid. The gas is turned on and the pressure maintained between 5 to 10 psi during mixing.
  • the solution is held after mixing for a 22 + 2 hour incubation.
  • the incubation is done at room temperature (20 - 25°C) and the in-process solution was protected from light.
  • the lipid siNA solution is diluted with an equal volume of Citrate buffer using a dual head peristaltic pump, Master Flex Peristaltic Pump, Model 7520-40 that is set up with equal lengths of tubing and a Tee connection and a flow rate of 360 mL/minute.
  • the ultrafiltration process is a timed process and the flow rates must be monitored carefully. This is a two step process; the first is a concentration step taking the diluted material from 32 liters to 3600 mLs and to a concentration of approximately 2 mg/mL. SIR-ONC-00001
  • a Flexstand with a ultrafiltration membrane GE PN UFP-100-C-35A installed is attached to the quatroflow pump. 200 mL of WFI is added to the reservoir followed by 3 liters of 0.5 N sodium hydroxide which is then flushed through the retentate to waste. This process is repeated three times. Then 3 L WFI are flushed through the system twice followed by 3 L of citrate buffer. The pump is then drained.
  • the diluted LNP solution is placed into the reservoir to the 4 liter mark.
  • the pump is turned on and the pump speed adjusted so the permeate flow rate is 300 mL/min. and the liquid level is constant at 4L in the reservoir.
  • the pump is stopped when all the diluted LNP solution has been transferred to the reservoir.
  • the diluted LNP solution is concentrated to 3600 mL in 240 minutes by adjusting the pump speed as necessary.
  • the second step is a diafiltration step exchanging the ethanol citrate buffer to phosphate buffered saline.
  • the diafiltration step takes 3 hours and again the flow rates must be carefully monitored. During this step, the ethanol concentration is monitored by head space GC. After 3 hours (20 diafiltration volumes), a second concentration is undertaken to concentrate the solution to approximately 6 mg/mL or a volume of 1.2 liters. This material is collected into a depyrogenated glass vessel. The system is rinsed with 400 mL of PBS at high flow rate and the permeate line closed. This material is collected and added to the first collection. The expected concentration at this point is 4.5 mg/mL. The concentration and volume are determined.
  • the feed tubing of the peristaltic pump is placed into a container containing 72 L of PBS (0.05 ⁇ filtered) and the flow rate is adjusted initially to maintain a constant volume of 3600 mL in the reservoir and then increased to 400 mL/min.
  • the LNP solution is diafiltered with PBS (20 volumes) for 180 minutes.
  • the LNP solution is concentrated to the 1.2 liter mark and collected into a depyrogenated 2 L graduated cylinder. 400 mL of PBS are added to the reservoir and the pump is recirculated for 2 minutes. The rinse is collected and added to the collected LNP solution in the graduated cylinder. SIR-ONC-00001
  • the obtained LNPs are characterized in terms of particle size, Zeta potential, alcohol content, total lipid content, nucleic acid encapsulated, and total nucleic acid concentration.
  • met proto-oncogene hepatocyte growth factor receptor
  • MET proto-oncogene
  • MET proto-oncogene
  • CAP any terminal cap, see for example Figure 5.
  • All Stab chemistries can comprise 3 '-overhang nucleotides having 2'-0-alkyl, 2'-deoxy-2'- fluoro, 2'-deoxy, LNA or other modified nucleotides or non-nucleotides.
  • All Stab chemistries typically comprise about 19-21 nucleotides, but can vary as described herein.
  • All Stab chemistries can also include a single ribonucleotide in the sense or passenger strand at the 11 th base paired position of the double- stranded nucleic acid duplex as determined from the 5 '-end of the antisense or guide strand (see Figure 4C).
  • Stab53*, Stab 53N*, and Stab 54 have three ribonucleotides at 5'-terminus.
  • ⁇ Stab 35 has 2'-0-methyl U at 3'-overhangs and three ribonucleotides at 5'-terminus ⁇ Stab 36 has 2'-0-methyl overhangs that are complementary to the target sequence
  • Stab35G*, Stab 35N*, Stab35rev*, Stab50*, Stab53*, and Stab53N* do not allow for a 2"-0- methyl modification at position 14 of the guide strand as determined from position the 5'- end.
  • Tandem synthesis utilizes double coupling of linker molecule
  • N/P ratio Nitrogen:Phosphorous ratio between cationic lipid and nucleic acid
  • the 2KPEG utilized is PEG2000, a polydispersion which can typically vary from -1500 to -3000 Da (i.e., where PEG(n) is about 33 to about 67, or on average -45).

Abstract

The present invention relates to compounds, compositions, and methods for the study, diagnosis, and treatment of traits, diseases and conditions that respond to the modulation of MET gene expression and/or activity, and/or modulate a MET gene expression pathway. Specifically, the invention relates to double-stranded nucleic acid molecules including small nucleic acid molecules, such as short interfering nucleic acid (siNA), short interfering RNA (siRNA), double-stranded RNA (dsRNA), micro-RNA (miRNA), and short hairpin RNA (shRNA) molecules that are capable of mediating or that mediate RNA interference (RNAi) against MET gene expression.

Description

SIR-ONC-00001
RNA INTERFERENCE MEDIATED INHIBITION OF MET GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
[0001] This application claims the benefit of U.S. Provisional Application No. 61/323,284 filed April 12, 2010. The above listed application is hereby incorporated by reference herein in its entirety, including the drawings.
SEQUENCE LISTING
[0002] The sequence listing submitted via EFS, in compliance with 37 CFR § 1.52(e)(5), is incorporated herein by reference. The sequence listing text file submitted via EFS contains the file "SequenceListing_SIRONCl", created on April 4, 2011, which is 721,386 bytes in size.
BACKGROUND OF THE INVENTION
[0003] Studies on signal transduction pathways have generated various promising molecular targets for therapeutic inhibition in cancer therapy. Receptor tyrosine kinases (RTK) represent an important class of such therapeutic targets. Recently, members of the MET proto-oncogene family, a subfamily of receptor tyrosine kinases, have drawn special attention to the association between invasion and metastasis. The MET family, including MET (also referred to as c-MET) and RON receptors, can function as oncogenes like most tyrosine kinases. MET has been shown to be overexpressed and/or mutated in a variety of malignancies. A number of MET activating mutations, many of which are located in the tyrosine kinase domain, have been detected in various solid tumors and have been implicated in invasion and metastasis of tumor cells.
[0004] The MET proto-oncogene encodes the MET receptor tyrosine kinase. The MET receptor is an approximately 190kDa glycosylated dimeric complex composed of a 50kDa alpha chain disulfide-linked to a 145kDa beta chain. The alpha chain is found extracellularly while the beta chain contains extracellular, transmembrane, and cytosolic domains. The c-MET protein is synthesized as a precursor and is proteolytically cleaved to yield mature alpha and beta subunits. It displays structural similarities to semaphoring and plexins, a ligand-receptor family that is involved in cell-cell interaction. SIR-ONC-00001
[0005] The natural ligand for MET is hepatocyte growth factor (HGF), a disulfide linked heterodimeric member of the scatter factor family that is produced predominantly by mesenchymal cells and acts primarily on MET-expressing epithelial and endothelial cells in an endocrine and/or paraendocrine fashion. HGF has some homology to plasminogen.
[0006] It is known that stimulation of MET via hepatocyte growth factor (also known as scatter factor, HGF/SF) results in a plethora of biological and biochemical effects in the cell. For example, binding of HGF to the MET extracellular domain results in receptor multimerization and phosphorylation of multiple tyrosine residues in the MET intracellular domain. Activation of MET signaling can lead to a wide array of cellular responses including proliferation, survival, angiogenesis, wound healing, tissue regeneration, scattering, motility, invasion and branching morphogenesis. HGF/MET signaling also plays a major role in the invasive growth that is found in most tissues, including cartilage, bone, blood vessels, and neurons.
[0007] Multiple reports have shown that MET is overexpressed in hepatocellular carcinomas (HCC) compared to normal liver tissue. Many studies have established a correlation between high MET expression and more aggressive HCC disease and reduced rate of patient survival. Ueki et al., Hepatology 1997 April 25 (4):862-6, reported a decrease in 5 year survival by -60% in patients with high MET expression compared to HCC patients with low MET expression.
[0008] Preclinical studies also provided ample evidence that activation of the MET pathway is important in initiation and progression of HCC. Zang et al., J. Mol. Cancer Ther. 2005 Oct.; 4(10): 1577-84, showed that the HCC cell line MHCC97-L, has high expression and constitutive activation of MET. Downregulation of MET expression by treating these cells with MET siRNA adenovirus inhibited their proliferation, colony forming ability, and tumorigenicity.
[0009] In another report, Wang et al. J. Cell Biol. 2001 May 28; 153(5): 1023-34, showed that transgenic mice overexpressing human MET develop HCC by 3 months of age and that these liver tumors regress when exogenous MET expression is turned off, suggesting that MET expression is critical for the maintenance in addition to initiation of these tumors. SIR-ONC-00001
[0010] In addition to its role in development and maintenance of liver tissue, MET and its ligand HGF have been implicated in progression of several types of human cancers including liver, prostate, colon, breast, and skin cancers.
[0011] Various MET mutations have been well described in multiple solid tumors and some hematologic malignancies. The prototypic MET mutation examples are seen in hereditary and sporadic human papillary renal carcinoma (Schmidt, L. et al., Nat. Tenet. 1997, 16, 68-73; Jeffers, M. et al, Proc. Nat. Acad. Sci. 1997, 94, 11445-11500). Other reported examples of MET mutations include ovarian cancer, childhood hepatocellular carcinoma, metastatic head and neck squamous cell carcinomas and gastric cancers. HGF/MET has been shown to inhibit anoikis, suspension- induced programmed cell death (apoptosis), in head and neck squamous cell carcinoma cells.
[0012] MET signaling is implicated in various cancers, especially renal. The nexus between MET and colorectal cancer has also been established. Analysis of MET expression during colorectal cancer progression showed that 50% of the carcinoma specimens analyzed expressed 5 to 50-fold higher levels of MET mRNA transcripts and protein versus the adjacent normal colonic mucosa. In addition, when compared to the primary tumor, 70% of colorectal cancer liver metastasis showed MET overexpression.
[0013] MET is also implicated in glioblastoma. High-grade malignant gliomas are the most common cancers of the central nervous system. Despite treatment with surgical resection, radiation therapy, and chemotherapy, the mean overall survival is < 1.5 years, and few patients survive for > 3 years. Human malignant gliomas frequently express both HGF and MET, which can establish an autocrine loop of biological significance. Glioma MET expression correlates with glioma grade, and an analysis of human tumor specimens showed that malignant gliomas have a 7-fold higher HGF content than low-grade gliomas. Multiple studies have demonstrated that human gliomas frequently co-express HGF and MET and that high levels of expression are associated with malignant progression. It was further shown that HGF-MET is able to activate Akt and protect glioma cell lines from apoptotic death, both in vitro and in vivo. SIR-ONC-00001
[0014] A number of reviews on MET and its function as an oncogene have been published: see for example Cancer and Metastasis Review 22:309-325 (2003); Nature Reviews/Molecular Cell Biology 4:915-925 (2003); Nature Reviews/Cancer 2:289-300 (2002).
[0015] Since dysregulation of the HGF/MET signaling has been implicated as a factor in tumorgenesis and disease progression in many tumors, different strategies for therapeutic inhibition of MET should be investigated. Specific inhibitors against MET gene expression have important therapeutic value for the treatment of cancers in which MET activity contributes to the invasive/metastatic phenotype. For this reason, there remains a need for molecules that inhibit MET.
[0016] Alteration of gene expression, specifically MET gene expression, through RNA interference (hereinafter "RNAi") is a one approach for meeting this need. RNAi is induced by short single-stranded RNA ("ssRNA") or double-stranded RNA ("dsRNA") molecules. The short dsRNA molecules, called "short interfering nucleic acids ("siNA")" or "short interfering RNA" or "siRNA" or "RNAi inhibitors" silence the expression of messenger RNAs ("mRNAs") that share sequence homology to the siNA. This can occur via cleavage of the mRNA mediated by an endonuclease complex containing a siNA, commonly referred to as an RNA-induced silencing complex (RISC). Cleavage of the target RNA typically takes place in the middle of the region complementary to the guide sequence of the siNA duplex (Elbashir et ah, 2001, Genes Dev., 15, 188). In addition, RNA interference can also involve small RNA {e.g., micro-RNA or miRNA) mediated gene silencing, presumably though cellular mechanisms that either inhibit translation or that regulate chromatin structure and thereby prevent transcription of target gene sequences (see for example Allshire, 2002, Science, 29 ', 1818-1819; Volpe et ah, 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al, 2002, Science, 297, 2232-2237). Despite significant advances in the field of RNAi, there remains a need for agents that can inhibit MET gene expression and that can treat disease associated with MET expression such as cancer. SIR-ONC-00001
SUMMARY OF THE INVENTION
[0017] The invention provides a solution to the problem of treating diseases that respond to the modulation of the MET gene expression using novel short interfering nucleic acid (siNA) molecules to modulate MET expression.
[0018] The present invention provides compounds, compositions, and methods useful for modulating the expression of MET genes, specifically those MET genes associated with cancer and for treating such conditions by RNA interference (RNAi) using small nucleic acid molecules.
[0019] In particular, the instant invention features small nucleic acid molecules, i.e., short interfering nucleic acid (siNA) molecules including, but not limited to, short interfering RNA (siRNA), double-stranded RNA (dsRNA), micro-RNA (miRNA), short hairpin RNA (shRNA) and circular RNA molecules and methods used to modulate the expression of MET genes and/or other genes involved in pathways of MET gene expression and/or activity.
[0020] In one aspect, the invention provides double-stranded short interfering nucleic acid (siNA) molecules that inhibit the expression of a MET gene in a cell or mammal, wherein the double-stranded siNAs comprise a sense and an antisense stand. The antisense strand comprises a sequence that is complementary to at least a part of an RNA associated with the expression of the MET gene. The sense strand comprises a sequence that is complementary to the antisense strand. In specific embodiments, the antisense strand comprises at least 15 nucleotides having sequence complementarity to a target sequence set forth in Table la. In other and/or in the same embodiments, the antisense strand comprises at least a 15 nucleotide sequence of one of the antisense sequences set forth in Table lb. In some embodiments, the sense strand comprises at least a 15 nucleotide sequence of a sense strand sequence as set forth in Table lb.
[0021] In certain embodiments of this aspect of the invention, double-stranded short interfering nucleic acid (siNA) molecules are provided wherein the antisense stand comprises a modified sequence as set forth in Table lc that has sequence complementarity to a target sequence of the invention. In some embodiments, the sense strand also comprises a modified sequence as set forth in Table lc. SIR-ONC-00001
[0022] In certain embodiments, the present invention provides a double- stranded short interfering nucleic acid (siNA) molecule that modulates the expression of MET, wherein the siNA comprises a sense strand and an antisense strand; each strand is independently 15 to 30 nucleotides in length; and the antisense strand comprises at least 15 nucleotides having sequence complementary to any of:
5'-GUGGCACUUUACUUACUUU-3' (SEQ ID NO: 1); 5'-UUUACUUACUUUAACUGGA-3' (SEQ ID NO: 43);
5'-CAAGCAUCUUCAGUUACCG-3' (SEQ ID NO: 51); or
5 ' - AGAAGGAAGUGUUU AAUAU-3' (SEQ ID NO: 53).
[0023] In some embodiments of the invention, the antisense strand of a siNA molecule comprises at least a 15 nucleotide sequence of:
5 ' - AAAGUAAGUAAAGUGCC AC-3' (SEQ ID NO: 1049);
5'-UCCAGUUAAAGUAAGUAAA-3' (SEQ ID NO: 1091); 5'-CGGUAACUGAAGAUGCUUG-3' (SEQ ID NO: 1099); or
5'-AUAUUAAACACUUCCUUCU-3' (SEQ ID NO: 1101).
[0024] In some embodiments, the sense strand of a siNA molecule of the invention comprises at least a 15 nucleotide sequence of:
5'-GUGGCACUUUACUUACUUU-3' (SEQ ID NO: 1);
5'-UUUACUUACUUUAACUGGA-3' (SEQ ID NO: 43);
5'-CAAGCAUCUUCAGUUACCG-3' (SEQ ID NO: 51); or
5 ' - AGAAGGAAGUGUUU AAUAU-3' (SEQ ID NO: 53)..
[0025] In some embodiments, a siNA molecule of the invention comprises any of:
5'-GUGGCACUUUACUUACUUU-3' (SEQ ID NO: 1) and 5'- AAAGUAAGUAAAGUGCCAC-3' (SEQ ID NO: 1049); or SIR-ONC-00001
5'-UUUACUUACUUUAACUGGA-3' (SEQ ID NO: 43) and 5'- UCCAGUUAAAGUAAGUAAA-3' (SEQ ID NO: 1091); or
5'-CAAGCAUCUUCAGUUACCG-3' (SEQ ID NO: 51) and 5'- CGGUAACUGAAGAUGCUUG-3' (SEQ ID NO: 1099); or
5' - AG A AGG A AGUGUUU A AU AU-31 (SEQ ID NO: 53) and 5'- AUAUUAAACACUUCCUUCU-31 (SEQ ID NO: 1101).
[0026] In some embodiments of the invention, all of the nucleotides of siNAs of the invention are unmodified. In other embodiments, one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) of the nucleotide positions independently in either one or both strands of an siNA molecule are modified. Modifications include nucleic acid sugar modifications, base modifications, backbone (internucleotide linkage) modifications, non-nucleotide modifications, and/or any combination thereof. In certain instances, purine and pyrimidine nucleotides are differentially modified. For example, purine and pyrimidine nucleotides can be differentially modified at the 2'- sugar position (i.e., at least one purine has a different modification from at least one pyrimidine in the same or different strand at the 2'-sugar position). In certain instances the purines are unmodified in one or both strands, while the pyrimidines in one or both strands are modified. In certain other instances, the pyrimidines are unmodified in one or both strands, while the purines in one or both strands are modified. In some instances, at least one modified nucleotide is a 2'-deoxy-2'-fluoro nucleotide, a 2'-deoxy nucleotide, or a 2'-0-alkyl nucleotide. In some instances, at least 5 or more of the pyrimidine nucleotides in one or both stands are either all 2'-deoxy-2'-fluoro or all 2'-0-methyl pyrimidine nucleotides. In some instances, at least 5 or more of the purine nucleotides in one or both stands are either all 2'-deoxy-2'-fluoro or all 2'-0-methyl purine nucleotides. In certain instances, wherein the siNA molecules comprise one or more modifications as described herein, the nucleotides at positions 1, 2, and 3 at the 5' end of the guide (antisense) strand are unmodified.
[0027] In certain embodiments, the siNA molecules of the invention have 3' overhangs of one, two, three, or four nucleotide(s) on one or both of the strands. In other embodiments, the siNA molecules lack overhangs (i.e., have blunt ends). Preferably, the siNA molecule has 3' overhangs of two nucleotides on both the sense and antisense strands. The overhangs can be SIR-ONC-00001 modified or unmodified. Examples of modified nucleotides in the overhangs include, but are not limited to, 2'-0-alkyl nucleotides, 2'-deoxy-2'-fluoro nucleotides, locked nucleic acid (LNA) nucleotides, or 2'-deoxy nucleotides. The overhang nucleotides in the antisense strand can comprise nucleotides that are complementary to nucleotides in the MET target sequence. Likewise, the overhangs in the sense stand can comprise nucleotides that are in the MET target sequence. In certain instances, the siNA molecules of the invention have two 3' overhang nucleotides on the antisense stand that are 2'-0-alkyl (e.g., 2'-0-methyl) nucleotides and two 3' overhang nucleotides on the sense stand that are 2'-deoxy nucleotides. In other instances, the siNA molecules of the invention have two 3' overhang nucleotides that are 2'-0-alkyl (e.g., 2'-0- methyl) nucleotides on both the antisense stand and on the sense stand. In certain embodiments, the 2'-0-alkyl nucleotides are 2'-0-methyl uridine nucleotides. In certain instances, the overhangs also comprise one or more phosphorothioate linkages between nucleotides of the overhang.
[0028] In some embodiments, the siNA molecules of the invention have caps (also referred to herein as "terminal caps." The cap can be present at the 5'-terminus (5'-cap) or at the 3'- terminus (3'-cap) or can be present on both termini, such as at the 5' and 3' termini of the sense strand of the siNA.
[0029] The siNA molecules of the invention when double stranded can be symmetric or asymmetric. Each strand of these double stranded siNAs independently can range in nucleotide length between 3 and 30 nucleotides. Generally, each strand of the siNA molecules of the invention is about 15 to 30 (i.e., about 19, 20, 21, 22, 23 or 24) nucleotides in length.
[0030] The siNA molecules of the invention, which are double stranded or have a duplex structure, independently comprise about 3 to about 30 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) base pairs. Generally, the duplex structure of siNAs of the invention is between 15 and 30, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 base pairs in length.
[0031] In certain embodiments, double-stranded short interfering nucleic acid (siNA) molecules are provided, wherein the molecule has a sense strand and an antisense strand and comprises formula (A): SIR-ONC-00001
B NX3 — (Ν)χ2 B -3
B (N)xi NX4 [Ν]χ5 -5'
(A) wherein, the upper strand is the sense strand and the lower strand is the antisense strand of the double- stranded nucleic acid molecule; wherein the antisense strand comprises at least a 15 nucleotide sequence of SEQ ID NO: 1049, SEQ ID NO: 1091, SEQ ID NO: 1099, or SEQ ID NO: 1101, and the sense strand comprises a sequence having complementarity to the antisense strand; each N is independently a nucleotide which is unmodified or chemically modified or a non-nucleotide; each B is a terminal cap that is present or absent;
(N) represents overhanging nucleotides, each of which is independently unmodified or chemically modified;
[N] represents nucleotides that are ribonucleotides; XI and X2 are independently integers from 0 to 4; X3 is an integer from 15 to 30; X4 is an integer from 9 to 30; and
X5 is an integer from 0 to 6, provided that the sum of X4 and X5 is 15-30.
[0032] In one embodiment, the invention features a double- stranded short interfering nucleic acid (siNA) of formula (A); wherein
(a) one or more pyrimidine nucleotides in Νχ4 positions are independently 2'-deoxy- 2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof; SIR-ONC-00001
(b) one or more purine nucleotides in Νχ4 positions are independently 2'-deoxy-2'- fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof;
(c) one or more pyrimidine nucleotides in Νχ3 positions are independently 2'-deoxy- 2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof; and
(d) one or more purine nucleotides in Νχ3 positions are independently 2'-deoxy-2'- fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides,
[0033] The present invention further provides compositions comprising the double-stranded nucleic acids molecules described herein with optionally a pharmaceutically acceptable carrier or diluent.
[0034] The administration of the composition can be carried out by known methods, wherein the nucleic acid is introduced into a desired target cell in vitro or in vivo.
[0035] Commonly used techniques for introduction of the nucleic acid molecules of the invention into cells, tissues, and organisms include the use of various carrier systems, reagents and vectors. Non-limiting examples of such carrier systems suitable for use in the present invention include conjugates, nucleic-acid-lipid particles, lipid nanoparticles (LNP), liposomes, lipoplexes, micelles, virosomes, virus like particles (VLP), nucleic acid complexes, and mixtures thereof.
[0036] The compositions of the invention can be in the form of an aerosol, dispersion, solution (e.g., an injectable solution), a cream, ointment, tablet, powder, suspension or the like. These compositions may be administered in any suitable way, e.g. orally, sublingually, buccally, parenterally, nasally, or topically. In some embodiments, the compositions are aerosolized and delivered via inhalation.
[0037] The molecules and compositions of the present invention have utility over a broad range of therapeutic applications. Accordingly another aspect of this invention relates to the use of the compounds and compositions of the invention in treating a subject. The invention thus provides a method for treating a subject, such as a human, suffering from a condition which is mediated by the action, or by the loss of action, of MET, wherein the method comprises SIR-ONC-00001 administering to the subject an effective amount of a double-stranded short interfering nucleic acid (siNA) molecule of the invention. In certain embodiments, the condition is cancer.
[0038] These and other aspects of the invention will be apparent upon reference to the following detailed description and attached figures. To that end, patents, patent applications, and other documents are cited throughout the specification to describe and more specifically set forth various aspects of this invention. Each of these references cited herein is hereby incorporated by reference in its entirety, including the drawings.
BRIEF DESCRIPTION OF THE DRAWINGS
[0039] Figure 1 shows a non-limiting proposed mechanistic representation of target RNA degradation involved in RNAi. Double-stranded RNA (dsRNA), which is generated by RNA- dependent RNA polymerase (RdRP) from foreign single-stranded RNA, for example viral, transposon, or other exogenous RNA, activates the DICER enzyme that in turn generates siNA duplexes. Alternately, synthetic or expressed siNA can be introduced directly into a cell by appropriate means. An active siNA complex forms that recognizes a target RNA, resulting in degradation of the target RNA by the RISC endonuclease complex or in the synthesis of additional RNA by RNA-dependent RNA polymerase (RdRP), which can activate DICER and result in additional siNA molecules, thereby amplifying the RNAi response.
[0040] Figure 2 shows non-limiting examples of chemically modified siNA constructs of the present invention using a generalized structure of a representative siNA duplex. The specific modifications shown in the figure can be utilized alone or in combination with other modifications of the figure, in addition to other modifications and features described herein with reference to any siNA molecule of the invention. In the figure, N stands for any nucleotide or optionally a non-nucleotide as described here. The upper strand, having Β-Νχ3-(Ν)χ2-Β -3' is the sense (or passenger) strand of the siNA, whereas the lower strand, having B(N)xi-Nx4-[N]x5 -5' is the antisense (or guide) strand of the siNA. Nucleotides (or optional non-nucleotides) of internal portions of the sense strand are designated Nxs and nucleotides (or optional non- nucleotides) of internal portions of the antisense strand are designated Νχ4. Nucleotides (or optional non-nucleotides) of the internal portions are generally base paired between the two strands, but can optionally lack base pairing (e.g. have mismatches or gaps) in some SIR-ONC-00001 embodiments. Nucleotides (or optional non-nucleotides) of overhang regions are designated by parenthesis (N). Nucleotides of the 5'-terminal portion of the antisense strand are designated [N]. Terminal caps are optionally present at the 5' and/or 3' end of the sense strand and further optionally present at the 3'-end of the antisense strand. Generally, each strand can independently range from about 15 to about 30 nucleotides in length, but can vary depending on the presence of any overhang nucleotides. In certain embodiments, XI and X2 are independently integers from 0 to 4; X3 is an integer from 15 to 30; X4 is an integer from 9 to 30; X5 is an integer from 0 to 6, provided that the sum of X4 and X5 is 15-30. Various modifications are shown for the nucleotides of the sense and antisense strands of the siNA constructs. The (N) overhang nucleotide positions can be chemically modified as described herein (e.g., 2'-0-methyl, 2'- deoxy-2'-fluoro, 2'-deoxy, LNA, universal bases etc.) and can be either derived from a corresponding target nucleic acid sequence or not. The constructs shown in the figure can also comprise phosphorothioate linkages as described herein. For example, phosphorothioate linkages can exist between any N, (N), and/or [N] positions. Such phosphorothioate incorporation can be utilized between purine "R" and pyrimidine "Y" positions, or for stabilization of pyrimidine linkages in general. Furthermore, although not depicted on the Figure, the constructs shown in the figure can optionally include a ribonucleotide at the 9th position from the 5'-end of the sense strand or the 11th position based on the 5'-end of the guide strand by counting 11 nucleotide positions in from the 5'-terminus of the guide strand. Similarly, the antisense strand can include a ribonucleotide at the 14th position from the 5'-end, or alternately can be selected or designed so that a 2'-0-alkyl nucleotide (e.g., a 2'-0-methyl purine) is not present at this position. Furthermore, although not shown in the Figure, the 5'-terminal position of the antisense strand can comprise a terminal phosphate group as described herein. The antisense strand generally comprises sequence complementary to any target nucleic acid sequence of the invention, such as those set forth in Table la herein.
[0041] Figure 3 shows non-limiting examples of certain combinations of modifications applied to the representative siNA duplex described in Figure 2. The table shown below the representative structure provides specific combinations of (Ν)χ1; (Ν)χ2, Νχ3, Νχ4, and/or [Ν]χ5 nucleotide (and optional non-nucleotide) positions. For example, combinations of 5 or more (e.g., 5, 6, 7, 8, 9, or 10 or more) Νχ3 and 5 or more (e.g., 5, 6, 7, 8, 9, or 10 or more) Νχ4 pyrimidine "Y" and purine "R" nucleotides are specified, each of which can independently have SIR-ONC-00001 specific (Ν)χ1 ; and/or (Ν)χ2, substitutions as shown in the figure, in addition to optional phosphorothioate substitutions. The 5'-terminal antisense strand [N] nucleotides are generally ribonucleotides, but can also be modified or unmodified depending on if they are purine "R" or pyrimidine "Y" nucleotides
[0042] Figure 4A-C shows non-limiting examples of different siNA constructs of the invention. The criteria of the representative structures shown in Figures 2 and 3 can be applied to any of the structures shown in Figure 4A-C.
[0043] The examples shown in Figure 4A (constructs 1, 2, and 3) have 19 representative base pairs; however, different embodiments of the invention include any number of base pairs described herein. Bracketed regions represent nucleotide overhangs, for example, comprising about 1, 2, 3, or 4 nucleotides in length, preferably about 2 nucleotides. Constructs 1 and 2 can be used independently for RNAi activity. Construct 2 can comprise a polynucleotide or non- nucleotide linker, which can optionally be designed as a biodegradable linker. In one embodiment, the loop structure shown in construct 2 can comprise a biodegradable linker that results in the formation of construct 1 in vivo and/or in vitro. In another example, construct 3 can be used to generate construct 2 under the same principle wherein a linker is used to generate the active siNA construct 2 in vivo and/or in vitro, which can optionally utilize another biodegradable linker to generate the active siNA construct 1 in vivo and/or in vitro. As such, the stability and/or activity of the siNA constructs can be modulated based on the design of the siNA construct for use in vivo or in vitro and/or in vitro.
[0044] The examples shown in Figure 4B represent different variations of double-stranded nucleic acid molecule of the invention, such as microRNA, that can include overhangs, bulges, loops, and stem-loops resulting from partial complementarity. Such motifs having bulges, loops, and stem-loops are generally characteristics of miRNA. The bulges, loops, and stem-loops can result from any degree of partial complementarity, such as mismatches or bulges of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more nucleotides in one or both strands of the double-stranded nucleic acid molecule of the invention.
[0045] The example shown in Figure 4C represents a model double- stranded nucleic acid molecule of the invention comprising a 19 base pair duplex of two 21 nucleotide sequences SIR-ONC-00001 having dinucleotide 3 '-overhangs. The top strand (1) represents the sense strand (passenger strand), the middle strand (2) represents the antisense (guide strand), and the lower strand (3) represents a target polynucleotide sequence. The dinucleotide overhangs (NN) can comprise a sequence derived from the target polynucleotide. For example, the 3'-(NN) sequence in the guide strand can be complementary to the 5'-[NN] sequence of the target polynucleotide. In addition, the 5'-(NN) sequence of the passenger strand can comprise the same sequence as the 5'-[NN] sequence of the target polynucleotide sequence. In other embodiments, the overhangs (NN) are not derived from the target polynucleotide sequence, for example where the 3'-(NN) sequence in the guide strand are not complementary to the 5'-[NN] sequence of the target polynucleotide and the 5'-(NN) sequence of the passenger strand can comprise different sequence from the 5'-[NN] sequence of the target polynucleotide sequence. In additional embodiments, any (NN) nucleotides are chemically modified, e.g., as 2'-0-methyl, 2'-deoxy-2'- fluoro, and/or other modifications herein. Furthermore, the passenger strand can comprise a ribonucleotide position N of the passenger strand. For the representative 19 base pair 21 mer duplex shown, position N can be 9 nucleotides in from the 3' end of the passenger strand. However, in duplexes of differing length, the position N is determined based on the 5'-end of the guide strand by counting 11 nucleotide positions in from the 5'-terminus of the guide strand and picking the corresponding base paired nucleotide in the passenger strand. Cleavage by Ago2 takes place between positions 10 and 11 as indicated by the arrow. In additional embodiments, there are two ribonucleotides, AW, at positions 10 and 11 based on the 5'-end of the guide strand by counting 10 and 11 nucleotide positions in from the 5'-terminus of the guide strand and picking the corresponding base paired nucleotides in the passenger strand.
[0046] Figure 5 shows non-limiting examples of different stabilization chemistries (1-10) that can be used, for example, to stabilize the 5' and/or 3'-ends of siNA sequences of the invention, including (1) [3-3'] -inverted deoxyribose; (2) deoxyribonucleotide; (3) [5'-3']-3'- deoxyribonucleotide; (4) [5'-3'] -ribonucleotide; (5) [5'-3']-3'-0-methyl ribonucleotide; (6) 3'- glyceryl; (7) [3'-5']-3'-deoxyribonucleotide; (8) [3'-3'] -deoxyribonucleotide; (9) [5 -2*]- deoxyribonucleotide; and (10) [5-3']-dideoxyribonucleotide (when X = O). In addition to modified and unmodified backbone chemistries indicated in the figure, these chemistries can be combined with different sugar and base nucleotide modifications as described herein. SIR-ONC-00001
[0047] Figure 6 shows a non-limiting example of a strategy used to identify chemically modified siNA constructs of the invention that are nuclease resistant while preserving the ability to mediate RNAi activity. Chemical modifications are introduced into the siNA construct based on educated design parameters (e.g. introducing 2'-modifications, base modifications, backbone modifications, terminal cap modifications etc). The modified construct is tested in an appropriate system (e.g., human serum for nuclease resistance, shown, or an animal model for PK/delivery parameters). In parallel, the siNA construct is tested for RNAi activity, for example in a cell culture system such as a luciferase reporter assay and/or against endogenous mRNA). Lead siNA constructs are then identified which possess a particular characteristic while maintaining RNAi activity, and can be further modified and assayed once again. This same approach can be used to identify siNA-conjugate molecules with improved pharmacokinetic profiles, delivery, and RNAi activity.
[0048] Figure 7 shows non-limiting examples of phosphorylated siNA molecules of the invention, including linear and duplex constructs and asymmetric derivatives thereof.
[0049] Figure 8 shows non-limiting examples of chemically modified terminal phosphate groups of the invention.
[0050] Figure 9 shows a non-limiting example of a cholesterol linked phosphoramidite that can be used to synthesize cholesterol conjugated siNA molecules of the invention. An example is shown with the cholesterol moiety linked to the 5' -end of the sense strand of an siNA molecule.
[0051] Figure 10 depicts an embodiment of 5' and 3' inverted abasic cap linked to a nucleic acid strand.
[0052] Figure 11 shows the efficacy of various MET siNAs in an LNP formulation (DLinDMA/Cholesterol/S-PEG-C-DMA/DSPC in a 40/48/2/10 ratio with an N/P ratio of 6) at 21 days using a dosing schedule of the 6mpk/week in TRE-MET mice.
[0053] Figure 12 shows the log2(fold change) in MET mRNA gene expression (reported as AACt) in Rhesus macaque monkeys at 3, 7, 14 and 28 days post-dosing with various MET siNAs in an LNP formulation (DLinDMA/Cholesterol/S-PEG-C-DMA/DSPC in a 40/48/2/10 ratio with SIR-ONC-00001 an N/P ratio of 6). The level of MET gene expression was also determined at 7 days pre-dosing. PBS and an ApoB siNA were used as controls.
DETAILED DESCRIPTION OF THE INVENTION
A. Terms and Definitions
[0054] The following terminology and definitions apply as used in the present application.
[0055] The term "abasic" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to sugar moieties lacking a nucleobase or having a hydrogen atom (H) or other non-nucleobase chemical groups in place of a nucleobase at the Γ position of the sugar moiety, see for example Adamic et ah, U.S. Pat. No. 5,998,203. In one embodiment, an abasic moiety of the invention is a ribose, deoxyribose, or dideoxyribose sugar.
[0056] The term "acyclic nucleotide" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to any nucleotide having an acyclic ribose sugar, for example where any of the ribose carbon/carbon or carbon/oxygen bonds are independently or in combination absent from the nucleotide.
[0057] The term "alkyl" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to a saturated or unsaturated hydrocarbons, including straight- chain, branched-chain, alkenyl, alkynyl groups and cyclic groups, but excludes aromatic groups. Notwithstanding the foregoing, alkyl also refers to non-aromatic heterocyclic groups. Preferably, the alkyl group has 1 to 12 carbons. More preferably, it is a lower alkyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkyl group can be substituted or unsubstituted. When substituted, the substituted group(s) is preferably, hydroxyl, halogen, cyano, C1-C4 alkoxy, =0, =S, N02 , SH, NH2, or NRiR2, where Ri and R2 independently are H or C1-C4 alkyl.
[0058] The phrase "agents that interfere with cell cycle checkpoints" refers to compounds that inhibit protein kinases that transduce cell cycle checkpoint signals, thereby sensitizing the cancer cell to DNA damaging agents. SIR-ONC-00001
[0059] The phrase "agents that interfere with receptor tyrosine kinases (RTKs)" refers to compounds that inhibit RTKs and therefore inhibit mechanisms involved in oncogenesis and tumor progression.
[0060] The phrase "androgen receptor modulators" refers to compounds that interfere or inhibit the binding of androgens to the receptor, regardless of mechanism.
[0061] The phrase "angiogenesis inhibitors" refers to compounds that inhibit the formation of new blood vessels, regardless of mechanism.
[0062] The term "aryl" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to an aromatic group that has at least one ring having a conjugated pi electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which can be optionally substituted. The preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, C1-C4 alkoxy, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, NH2, and NRiR2 groups, where R and R2 independently are H or C1-C4 alkyl.
[0063] The term "alkylaryl" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to an alkyl group (as described above) covalently joined to an aryl group (as described above). Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted. Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen, and examples of heterocyclic aryl groups having such heteroatoms include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted. Preferably, the alkyl group is a C1-C4 alkyl group.
[0064] The term "amide" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to an -C(0)-NH-R, where R is either alkyl, aryl, alkylaryl or hydrogen.
[0065] The phrase "antisense region" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the SIR-ONC-00001 term refers to a nucleotide sequence of an siNA molecule having complementarity to a target nucleic acid sequence. In addition, the antisense region of an siNA molecule can optionally comprise a nucleic acid sequence having complementarity to a sense region of the siNA molecule. In one embodiment, the antisense region of the siNA molecule is referred to as the antisense strand or guide strand.
[0066] The phrase "asymmetric hairpin" refers to a linear siNA molecule comprising an antisense region, a loop portion that can comprise nucleotides or non-nucleotides, and a sense region that comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complementary nucleotides to base pair with the antisense region and form a duplex with loop. For example, an asymmetric hairpin siNA molecule of the invention can comprise an antisense region having length sufficient to mediate RNAi in a cell or in vitro system {e.g. about 15 to about 30, or about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides) and a loop region comprising about 4 to about 12 {e.g. , about 4, 5, 6, 7, 8, 9, 10, 11, or 12) nucleotides, and a sense region having about 3 to about 25 {e.g. , about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides that are complementary to the antisense region. The asymmetric hairpin siNA molecule can also comprise a 5 '-terminal phosphate group that can be chemically modified. The loop portion of the asymmetric hairpin siNA molecule can comprise nucleotides, non-nucleotides, linker molecules, or conjugate molecules as described herein.
[0067] The term "biodegradable" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to degradation in a biological system, for example, enzymatic degradation or chemical degradation.
[0068] The term "biodegradable linker" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a linker molecule that is designed to connect one molecule to another molecule, and which is susceptible to degradation in a biological system. The linker can be a nucleic acid or non-nucleic acid based linker. For example, a biodegradable linker can be used to attach a ligand or biologically active molecule to an siNA molecule of the invention. Alternately, a biodegradable linker can be used to connect the sense and antisense strands of an siNA molecule SIR-ONC-00001 of the invention. The biodegradable linker is designed such that its stability can be modulated for a particular purpose, such as delivery to a particular tissue or cell type. The stability of a nucleic acid-based biodegradable linker molecule can be modulated by using various chemistries, for example combinations of ribonucleotides, deoxyribonucleotides, and chemically modified nucleotides, such as 2'-0-methyl, 2'-fluoro, 2'-amino, 2'-0-amino, 2'-C-allyl, 2'-0-allyl, and other 2'-modified or base modified nucleotides. The biodegradable nucleic acid linker molecule can be a dimer, trimer, tetramer or longer nucleic acid molecule, for example, an oligonucleotide of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides in length, or can comprise a single nucleotide with a phosphorus-based linkage, for example, a phosphoramidate or phosphodiester linkage. The biodegradable nucleic acid linker molecule can also comprise nucleic acid backbone, nucleic acid sugar, or nucleic acid base modifications.
[0069] The phrase "biologically active molecule" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to compounds or molecules that are capable of eliciting or modifying a biological response in a system and/or are capable of modulating the pharmacokinetics and/or pharmacodynamics of other biologically active molecules. Examples of biologically active molecules, include siNA molecules alone or in combination with other molecules including, but not limited to therapeutically active molecules such as antibodies, cholesterol, hormones, antivirals, peptides, proteins, chemotherapeutics, small molecules, vitamins, co-factors, nucleosides, nucleotides, oligonucleotides, enzymatic nucleic acids, antisense nucleic acids, triplex forming oligonucleotides, polyamines, polyamides, polyethylene glycol, other polyethers, 2-5A chimeras, siNA, dsRNA, allozymes, aptamers, decoys and analogs thereof.
[0070] The phrase "biological system" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to material, in a purified or unpurified form, from biological sources including, but not limited to, human or animal, wherein the system comprises the components required for RNAi activity. Thus, the phrase includes, for example, a cell, tissue, subject, or organism, or extract thereof. The term also includes reconstituted material from a biological source. SIR-ONC-00001
[0071] The phrase "blunt end" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to termini of a double-stranded siNA molecule having no overhanging nucleotides. For example, the two strands of a double- stranded siNA molecule having blunt ends align with each other with matched base-pairs without overhanging nucleotides at the termini. A siNA duplex molecule of the invention can comprise blunt ends at one or both termini of the duplex, such as termini located at the 5'-end of the antisense strand, the 5'-end of the sense strand, or both termini of the duplex.
[0072] The term "cap" also referred to herein as "terminal cap," as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a moiety, which can be a chemically modified nucleotide or non-nucleotide that can be incorporated at one or more termini of one or more nucleic acid molecules of the invention. These terminal modifications protect the nucleic acid molecule from exonuclease degradation, and can help in delivery and/or localization within a cell. The cap can be present at the 5'-terminus (5'-cap) or at the 3'-terminal (3'-cap) or can be present on both termini of any nucleic acid molecule of the invention. A cap can be present at the 5'-end, 3-end and/or 5' and 3'-ends of the sense strand of a nucleic acid molecule of the invention. Additionally, a cap can optionally be present at the 3'-end of the antisense strand of a nucleic acid molecule of the invention. In non-limiting examples, the 5'-cap includes, but is not limited to, LNA; glyceryl; inverted deoxy abasic residue (moiety); 4',5'-methylene nucleotide; l-(beta- D-erythrofuranosyl) nucleotide, 4'-thio nucleotide; carbocyclic nucleotide; 1,5-anhydrohexitol nucleotide; L-nucleotides; alpha-nucleotides; modified base nucleotide; phosphorodithioate linkage; ί/ireopentofuranosyl nucleotide; acyclic 3',4'-seco nucleotide; acyclic 3,4- dihydroxybutyl nucleotide; acyclic 3,5-dihydroxypentyl nucleotide; 3'-3'-inverted nucleotide moiety; 3'-3'-inverted abasic moiety; 3'-2'-inverted nucleotide moiety; 3'-2'-inverted abasic moiety; 1,4-butanediol phosphate; 3'-phosphoramidate; hexylphosphate; aminohexyl phosphate; 3'-phosphate; 3'-phosphorothioate; phosphorodithioate; or bridging or non-bridging methylphosphonate moiety. Non-limiting examples of the 3'-cap include, but are not limited to, LNA; glyceryl; inverted deoxy abasic residue (moiety); 4', 5'-methylene nucleotide; l-(beta-D- erythrofuranosyl) nucleotide; 4'-thio nucleotide; carbocyclic nucleotide; 5'-amino-alkyl phosphate; l,3-diamino-2-propyl phosphate; 3 -aminopropyl phosphate; 6-aminohexyl phosphate; SIR-ONC-00001
1,2-aminododecyl phosphate; hydroxypropyl phosphate; 1,5-anhydrohexitol nucleotide; L- nucleotide; alpha-nucleotide; modified base nucleotide; phosphorodithioate; threo- pentofuranosyl nucleotide; acyclic 3',4'-seco nucleotide; 3,4-dihydroxybutyl nucleotide; 3,5- dihydroxypentyl nucleotide, 5'-5'-inverted nucleotide moiety; 5'-5'-inverted abasic moiety; 5'- phosphoramidate; 5'-phosphorothioate; 1,4-butanediol phosphate; 5'-amino; bridging and/or non- bridging 5'-phosphoramidate; phosphorothioate and/or phosphorodithioate; bridging or non bridging methylphosphonate; and 5'-mercapto moieties (for more details see Beaucage and Iyer, 1993, Tetrahedron 49, 1925; incorporated by reference herein). Figure 5 shows some non- limiting examples of various caps.
[0073] The term "cell" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term is used in its usual biological sense, and does not refer to an entire multicellular organism, e.g., specifically does not refer to a human being. The cell can be present in an organism, e.g., birds, plants and mammals, such as humans, cows, sheep, apes, monkeys, swine, dogs, and cats. The cell can be prokaryotic {e.g., bacterial cell) or eukaryotic {e.g., mammalian or plant cell). The cell can be of somatic or germ line origin, totipotent or pluripotent, dividing or non-dividing. The cell can also be derived from or can comprise a gamete or embryo, a stem cell, or a fully differentiated cell.
[0074] The phrase "chemical modification" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to any modification of the chemical structure of the nucleotides that differs from nucleotides of native siRNA or RNA in general. The term "chemical modification" encompasses the addition, substitution, or modification of native siRNA or RNA at the sugar, base, or internucleotide linkage, as described herein or as is otherwise known in the art. In certain embodiments, the term "chemical modification" can refer to certain forms of RNA that are naturally occurring in certain biological systems, for example 2'-0-methyl modifications or inosine modifications.
[0075] The term "MET" refers to mesenchymal-epithelial transition factor which is a proto- oncogene that encodes MET proteins, MET peptides, MET polypeptides, MET regulatory polynucleotides (e.g., MET miRNAs and siNAs), mutant MET genes, and splice variants of a SIR-ONC-00001
MET genes, as well as other genes involved in MET pathways of gene expression and/or activity. It is also known as the hepatocyte growth factor receptor (HGFR). Thus, each of the embodiments described herein with reference to the term "MET" are applicable to all of the protein, peptide, polypeptide, and/or polynucleotide molecules covered by the term "MET", as that term is defined herein. Comprehensively, such gene targets are also referred to herein generally as "target" sequences (including Table 10).
[0076] The term "complementarity" or "complementary" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the terms generally refer to the formation or existence of hydrogen bond(s) between one nucleic acid sequence and another nucleic acid sequence by either traditional Watson-Crick or other non-traditional types of bonding as described herein. In reference to the nucleic molecules of the present invention, the binding free energy for a nucleic acid molecule with its complementary sequence is sufficient to allow the relevant function of the nucleic acid to proceed, e.g., RNAi activity. Determination of binding free energies for nucleic acid molecules is well known in the art (see, e.g., Turner et al., 1987, CSH Symp. Quant. Biol. LII pp.123- 133; Frier et al, 1986, Proc. Nat. Acad. Sci. USA 83:9373-9377; Turner et al, 1987, J. Am. Chem. Soc. 109:3783-3785). Perfect complementary means that all the contiguous residues of a nucleic acid sequence will hydrogen bond with the same number of contiguous residues in a second nucleic acid sequence. Partial complementarity can include various mismatches or non-based paired nucleotides (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more mismatches, non-nucleotide linkers, or non-based paired nucleotides) within the nucleic acid molecule, which can result in bulges, loops, or overhangs that result between the sense strand or sense region and the antisense strand or antisense region of the nucleic acid molecule or between the antisense strand or antisense region of the nucleic acid molecule and a corresponding target nucleic acid molecule. Such partial complementarity can be represented by a % complementarity that is determined by the number of non-base paired nucleotides, i.e., about 50%, 60%, 70%, 80%, 90% etc. depending on the total number of nucleotides involved. Such partial complementarity is permitted to the extent that the nucleic acid molecule (e.g., siNA) maintains its function, for example the ability to mediate sequence specific RNAi. SIR-ONC-00001
[0077] The terms "composition" or "formulation" as used herein refer to their generally accepted meaning in the art. These terms generally refer to a composition or formulation, such as in a pharmaceutically acceptable carrier or diluent, in a form suitable for administration, e.g., systemic or local administration, into a cell or subject, including, for example, a human. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, inhalation, or by injection. Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged nucleic acid is desirable for delivery). For example, compositions injected into the blood stream should be soluble. Other factors are known in the art, and include considerations such as toxicity and forms that prevent the composition or formulation from exerting its effect. As used herein, pharmaceutical formulations include formulations for human and veterinary use. Non-limiting examples of agents suitable for formulation with the nucleic acid molecules of the instant invention include: Lipid Nanoparticles (see for example Semple et al., 2010, Nat Biotechnol., Feb;28(2): 172-6.); P- glycoprotein inhibitors (such as Pluronic P85); biodegradable polymers, such as poly (DL- lactide-coglycolide) microspheres for sustained release delivery (Emerich, DF et al, 1999, Cell Transplant, 8, 47-58); and loaded nanoparticles, such as those made of polybutylcyanoacrylate. Other non-limiting examples of delivery strategies for the nucleic acid molecules of the instant invention include material described in Boado et al., 1998, J. Pharm. Set, 87, 1308-1315; Tyler et al, 1999, FEBS Lett., 421, 280-284; Pardridge et al., 1995, PNAS USA., 92, 5592-5596; Boado, 1995, Adv. Drug Delivery Rev., 15, 73-107; Aldrian-Herrada et al., 1998, Nucleic Acids Res., 26, 4910-4916; and Tyler et al., 1999, PNAS USA., 96, 7053-7058. A "pharmaceutically acceptable composition" or "pharmaceutically acceptable formulation" can refer to a composition or formulation that allows for the effective distribution of the nucleic acid molecules of the instant invention to the physical location most suitable for their desired activity.
[0078] The phrase "cytotoxic/cytostatic agents" refer to compounds that cause cell death or inhibit cell proliferation primarily by interfering directly with the cell's functioning or inhibit or interfere with cell mytosis, including alkylating agents, tumor necrosis factors, intercalators, hypoxia activatable compounds, microtubule inhibitors/micro tubule- stabilizing agents, inhibitors of mitotic kinesins, inhibitors of histone deacetylase, inhibitors of kinases involved in mitotic progression, antimetabolites; biological response modifiers; hormonal/anti-hormonal therapeutic SIR-ONC-00001 agents, hematopoietic growth factors, monoclonal antibody targeted therapeutic agents, topoisomerase inhibitors, proteasome inhibitors and ubiquitin ligase inhibitors.
[0079] The phrase "estrogen receptor modulators" refers to compounds that interfere with or inhibit the binding of estrogen to the receptor, regardless of mechanism.
[0080] The term "gene" or "target gene" as used herein refers to their meaning as is generally accepted in the art. The terms generally refer a nucleic acid (e.g., DNA or RNA) sequence that comprises partial length or entire length coding sequences necessary for the production of a polypeptide. The target gene can also include the UTR or non-coding region of the nucleic acid sequence. A gene or target gene can also encode a functional RNA (fRNA) or non-coding RNA (ncRNA), such as small temporal RNA (stRNA), micro RNA (miRNA), small nuclear RNA (snRNA), short interfering RNA (siRNA), small nucleolar RNA (snRNA), ribosomal RNA (rRNA), transfer RNA (tRNA) and precursor RNAs thereof. Such non-coding RNAs can serve as target nucleic acid molecules for siNA mediated RNA interference in modulating the activity of fRNA or ncRNA involved in functional or regulatory cellular processes. Aberrant fRNA or ncRNA activity leading to disease can therefore be modulated by siNA molecules of the invention. siNA molecules targeting fRNA and ncRNA can also be used to manipulate or alter the genotype or phenotype of a subject, organism or cell, by intervening in cellular processes such as genetic imprinting, transcription, translation, or nucleic acid processing (e.g., transamination, methylation etc.). The target gene can be a gene derived from a cell, an endogenous gene, a transgene, or exogenous genes such as genes of a pathogen, for example a virus, which is present in the cell after infection thereof. The cell containing the target gene can be derived from or contained in any organism, for example a plant, animal, protozoan, virus, bacterium, or fungus. Non-limiting examples of plants include monocots, dicots, or gymnosperms. Non-limiting examples of animals include vertebrates or invertebrates. Non- limiting examples of fungi include molds or yeasts. For a review, see for example Snyder and Gerstein, 2003, Science, 300, 258-260.
[0081] The phrase "HMG-CoA reductase inhibitors" refers to inhibitors of 3-hydroxy-3- methylglutaryl-CoA reductase. The term HMG-CoA reductase inhibitor as used herein includes all pharmaceutically acceptable lactone and open-acid forms (i.e., where the lactone ring is SIR-ONC-00001 opened to form the free acid) as well as salt and ester forms of compounds that have HMG-CoA reductase inhibitory activity, and therefore the use of such salts, esters, open-acid and lactone forms is included within the scope of this invention.
[0082] The phrase "homologous sequence" as used herein refers to its meaning as is generally accepted in the art. The term generally refers a nucleotide sequence that is shared by one or more polynucleotide sequences, such as genes, gene transcripts and/or non-coding polynucleotides. For example, a homologous sequence can be a nucleotide sequence that is shared by two or more genes encoding related but different proteins, such as different members of a gene family, different protein epitopes, different protein isoforms or completely divergent genes. A homologous sequence can be a nucleotide sequence that is shared by two or more non- coding polynucleotides, such as noncoding DNA or RNA, regulatory sequences, introns, and sites of transcriptional control or regulation. Homologous sequences can also include sequence regions shared by more than one polynucleotide sequence. Homology does not need to be perfect identity (100%), as partially homologous sequences are also contemplated by and within the scope of the instant invention {e.g., at least 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% etc.). Percent homology is the number of matching nucleotides between two sequences divided by the total length being compared, multiplied by 100.
[0083] The phrase "improved RNAi activity" refers to an increase in RNAi activity measured in vitro and/or in vivo, where the RNAi activity is a reflection of both the ability of the siNA to mediate RNAi and the stability of the siNAs of the invention. In this invention, the product of these activities can be increased in vitro and/or in vivo compared to an all RNA siNA or an siNA containing a plurality of ribonucleotides. In some cases, the activity or stability of the siNA molecule can be decreased {i.e., less than ten-fold), but the overall activity of the siNA molecule is enhanced in vitro and/or in vivo.
[0084] The terms "inhibit," "down-regulate," or "reduce" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, he term generally refers the reduction in the expression of the gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or SIR-ONC-00001 activity of one or more proteins or protein subunits, below that observed in the absence of the nucleic acid molecules (e.g. , siNA) of the invention. Down-regulation can also be associated with post-transcriptional silencing, such as, RNAi mediated cleavage or by alteration in DNA methylation patterns or DNA chromatin structure. Inhibition, down-regulation or reduction with an siNA molecule can be in reference to an inactive molecule, an attenuated molecule, an siNA molecule with a scrambled sequence, or an siNA molecule with mismatches or alternatively, it can be in reference to the system in the absence of the nucleic acid.
[0085] The phrase "inhibitors of cell proliferation and survival signaling pathway" refers to pharmaceutical agents that inhibit cell surface receptors and signal transduction cascades downstream of those surface receptors.
[0086] The term "integrin blockers" refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the ακβ3 integrin, to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the κβ5 integrin, to compounds which antagonize, inhibit or counteract binding of a physiological ligand to both the ακβ3 integrin and the κβ5 integrin, and to compounds which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells. The term also refers to antagonists of the
Figure imgf000028_0001
α2βι α5βι α6βι and α6β4 integrins. The term also refers to antagonists of any combination of ακβ3, κβ5,
Figure imgf000028_0002
α2βι α5βι α6βι and α6β4 integrins.
[0087] The terms "intermittent" or "intermittently" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to periodic stopping and starting at either regular or irregular intervals.
[0088] The terms "internucleoside linkage" or "internucleoside linker" or "internucleotide linkage" or "internucleotide linker" are used herein interchangeably and refer to any linker or linkage between two nucleoside units, as is known in the art, including, for example, but not limitation, phosphate, analogs of phosphate, phosphonate, guanidium, hydroxylamine, hydroxylhydrazinyl, amide, carbamate, alkyl, and substituted alkyl linkages. The internucleoside linkages constitute the backbone of a nucleic acid molecule. SIR-ONC-00001
[0089] The terms "mammalian" or "mammal" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to any warm blooded vertebrate species, such as a human, mouse, rat, dog, cat, hamster, guinea pig, rabbit, livestock, and the like.
[0090] The phrase "metered dose inhaler" or MDI refers to a unit comprising a can, a secured cap covering the can and a formulation metering valve situated in the cap. MDI systems includes a suitable channeling device. Suitable channeling devices comprise for example, a valve actuator and a cylindrical or cone-like passage through which medicament can be delivered from the filled canister via the metering valve to the nose or mouth of a patient such as a mouthpiece actuator.
[0091] The term "microRNA" or "miRNA" as used herein refers to its meaning as is generally accepted in the art. The term generally refers a small double- stranded RNA that regulates the expression of target messenger RNAs either by mRNA cleavage, translational repression/inhibition or heterochromatic silencing (see for example Ambros, 2004, Nature, 431, 350-355; Bartel, 2004, Cell, 116, 281-297; Cullen, 2004, Virus Research., 102, 3-9; He et al, 2004, Nat. Rev. Genet., 5, 522-531; Ying et al, 2004, Gene, 342, 25-28; and Sethupathy et al, 2006, RNA, 12: 192-197).
[0092] The term "modulate" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to when the expression of a gene, or level of one or more RNA molecules (coding or non-coding), or activity of one or more RNA molecules or proteins or protein subunits, is up-regulated or down-regulated, such that expression, level, or activity is greater than or less than that observed in the absence of the molecule that effects modulation. For example, the term "modulate" in some embodiments can refer to inhibition and in other embodiments can refer to potentiation or up-regulation, e.g., of gene expression.
[0093] The phrase "modified nucleotide" as used herein refers to its meaning as is generally accepted in the art. The term generally refers a nucleotide, which contains a modification in the chemical structure of the base, sugar and/or phosphate of the unmodified (or natural) nucleotide as is generally known in the art. Non-limiting examples of modified nucleotides are described herein and in U.S. Application No. 12/064,014. SIR-ONC-00001
[0094] The phrase "NSAIDs that are selective COX-2 inhibitors" for purposes herein, refers to NSAIDs, which possess a specificity for inhibiting COX-2 over COX-1 of at least 100 fold as measured by the ratio of IC50 for COX-2 over IC50 for COX-1 evaluated by cell or microsomal assays.
[0095] The phrase "non-base paired" refers to nucleotides that are not base paired between the sense strand or sense region and the antisense strand or antisense region of an double- stranded siNA molecule; and can include for example, but not limitation, mismatches, overhangs, single stranded loops, etc.
[0096] The term "non-nucleotide" refers to any group or compound which can be incorporated into a nucleic acid chain in the place of one or more nucleotide units, such as for example but not limitation abasic moieties or alkyl chains. The group or compound is "abasic" in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine and therefore lacks a nucleobase at the 1 '-position.
[0097] The term "nucleotide" is used as is generally recognized in the art. Nucleotides generally comprise a nucleobase, a sugar, and an internucleoside linkage, e.g., a phosphate. The base can be a natural bases (standard), modified bases, or a base analog, as are well known in the art. Such bases are generally located at the Γ position of a nucleotide sugar moiety. Additionally, the nucleotides can be unmodified or modified at the sugar, internucleoside linkage, and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and others; see, for example, U.S. Application No. 12/064,014.
[0098] The term "overhang" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary double stranded nucleic acid molecules, the term generally refers to the terminal portion of a nucleotide sequence that is not base paired between the two strands of a double- stranded nucleic acid molecule (see for example, Figure 4). Overhangs, when present, are typically at the 3'-end of one or both strands in a siNA duplex.
[0099] The term "parenteral" as used herein refers to its meaning as is generally accepted in the art. The term generally refers methods or techniques of administering a molecule, drug, SIR-ONC-00001 agent, or compound in a manner other than through the digestive tract, and includes epicutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, or intrathecal injection or infusion techniques and the like.
[0100] The phrase "pathway target" refers to any target involved in pathways of gene expression or activity. For example, any given target can have related pathway targets that can include upstream, downstream, or modifier genes in a biologic pathway. These pathway target genes can provide additive or synergistic effects in the treatment of diseases, conditions, and traits herein.
[0101] The term "phosphorothioate" refers to an intemucleotide phosphate linkage comprising one or more sulfur atoms in place of an oxygen atom. Hence, the term phosphorothioate refers to both phosphorothioate and phosphorodithioate intemucleotide linkages.
[0102] "Prenyl-protein transferase inhibitor" refers to a compound that inhibits any one or any combination of the prenyl-protein transferase enzymes, including farnesyl-protein transferase (FPTase), geranylgeranyl-protein transferase type I (GGPTase-I), and geranylgeranyl-protein transferase type-II (GGPTase-II, also called Rab GGPTase).
[0103] The phrase "retinoid receptor modulators" refers to compounds that interfere or inhibit the binding of retinoids to the receptor, regardless of mechanism.
[0104] The term "ribonucleotide" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to a nucleotide with a hydroxyl group at the 2' position of a β-D-ribofuranose moiety.
[0105] The term "RNA" as used herein refers to its generally accepted meaning in the art. Generally, the term RNA refers to a molecule comprising at least one ribofuranoside moiety. The term can include double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides. Such alterations can include addition of non-nucleotide material, such as to the end(s) of the siNA or internally, for example at one or SIR-ONC-00001 more nucleotides of the RNA. Nucleotides in the RNA molecules of the instant invention can also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.
[0106] The phrase "RNA interference" or term "RNAi" refer to the biological process of inhibiting or down regulating gene expression in a cell, as is generally known in the art, and which is mediated by short interfering nucleic acid molecules, see for example Zamore and Haley, 2005, Science, 309, 1519-1524; Vaughn and Martienssen, 2005, Science, 309, 1525-1526; Zamore et al, 2000, Cell, 101, 25-33; Bass, 2001, Nature, 411, 428-429; Elbashir et al, 2001, Nature, 411, 494-498; and Kreutzer et al, International PCT Publication No. WO 00/44895; Zernicka-Goetz et al., International PCT Publication No. WO 01/36646; Fire, International PCT Publication No. WO 99/32619; Plaetinck et al, International PCT Publication No. WO 00/01846; Mello and Fire, International PCT Publication No. WO 01/29058; Deschamps- Depaillette, International PCT Publication No. WO 99/07409; and Li et al, International PCT Publication No. WO 00/44914; Allshire, 2002, Science, 297, 1818-1819; Volpe et al, 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al, 2002, Science, 297 ', 2232-2237; Hutvagner and Zamore, 2002, Science, 297 ', 2056-60; McManus et al, 2002, RNA, 8, 842-850; Reinhart et al, 2002, Gene & Dev., 16, 1616-1626; and Reinhart & Bartel, 2002, Science, 297, 1831). Additionally, the term RNAi is meant to be equivalent to other terms used to describe sequence specific RNA interference, such as post transcriptional gene silencing, translational inhibition, transcriptional inhibition, or epigenetics. For example, siNA molecules of the invention can be used to epigenetically silence genes at either the post- transcriptional level or the pre-transcriptional level. In a non-limiting example, epigenetic modulation of gene expression by siNA molecules of the invention can result from siNA mediated modification of chromatin structure or methylation patterns to alter gene expression (see, for example, Verdel et al, 2004, Science, 303, 672-676; Pal-Bhadra et al, 2004, Science, 303, 669-672; Allshire, 2002, Science, 297, 1818-1819; Volpe et al, 2002, Science, 297, 1833- 1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al, 2002, Science, 297, 2232-2237). In another non-limiting example, modulation of gene expression by siNA molecules of the invention can result from siNA mediated cleavage of RNA (either coding or non-coding RNA) via RISC, or via translational inhibition, as is known in the art or modulation can result from SIR-ONC-00001 transcriptional inhibition (see for example Janowski et al, 2005, Nature Chemical Biology, 1, 216-222).
[0107] The phrase "RNAi inhibitor" refers to any molecule that can down regulate, reduce or inhibit RNA interference function or activity in a cell or organism. An RNAi inhibitor can down regulate, reduce or inhibit RNAi (e.g., RNAi mediated cleavage of a target polynucleotide, translational inhibition, or transcriptional silencing) by interaction with or interfering with the function of any component of the RNAi pathway, including protein components such as RISC, or nucleic acid components such as miRNAs or siRNAs. A RNAi inhibitor can be an siNA molecule, an antisense molecule, an aptamer, or a small molecule that interacts with or interferes with the function of RISC, a miRNA, or an siRNA or any other component of the RNAi pathway in a cell or organism. By inhibiting RNAi (e.g., RNAi mediated cleavage of a target polynucleotide, translational inhibition, or transcriptional silencing), a RNAi inhibitor of the invention can be used to modulate (e.g., up-regulate or down regulate) the expression of a target gene.
[0108] The phrase "sense region" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to a nucleotide sequence of an siNA molecule having complementarity to an antisense region of the siNA molecule. In addition, the sense region of an siNA molecule can comprise a nucleic acid sequence having homology or sequence identity with a target nucleic acid sequence. In one embodiment, the sense region of the siNA molecule is also referred to as the sense strand or passenger strand.
[0109] The phrases "short interfering nucleic acid", "siNA", "short interfering RNA", "siRNA", "short interfering nucleic acid molecule", "short interfering oligonucleotide molecule", or "chemically modified short interfering nucleic acid molecule" refer to any nucleic acid molecule capable of inhibiting or down regulating gene expression or viral replication by mediating RNA interference ("RNAi") or gene silencing in a sequence- specific manner. These terms can refer to both individual nucleic acid molecules, a plurality of such nucleic acid molecules, or pools of such nucleic acid molecules. The siNA can be a double-stranded nucleic acid molecule comprising self-complementary sense and antisense strands, wherein the antisense SIR-ONC-00001 strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense strand comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. The siNA can be a polynucleotide with a duplex, asymmetric duplex, hairpin or asymmetric hairpin secondary structure, having self-complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence in a separate target nucleic acid molecule or a portion thereof and the sense region comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. The siNA can be a circular single-stranded polynucleotide having two or more loop structures and a stem comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region comprises a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof, and wherein the circular polynucleotide can be processed either in vivo or in vitro to generate an active siNA molecule capable of mediating RNAi. The siNA can also comprise a single-stranded polynucleotide having a nucleotide sequence complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof (for example, where such siNA molecule does not require the presence within the siNA molecule of a nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof), wherein the single-stranded polynucleotide can further comprise a terminal phosphate group, such as a 5'-phosphate (see for example, Martinez et ah, 2002, Cell, 110, 563-574 and Schwarz et al., 2002, Molecular Cell, 10, 537-568), or 5',3'- diphosphate.
[0110] The term "subject" as used herein refers to its meaning as is generally accepted in the art. The term generally refers an organism to which the nucleic acid molecules of the invention can be administered. A subject can be a mammal or mammalian cells, including a human or human cells. The term also refers to an organism, which is a donor or recipient of explanted cells or the cells themselves.
[0111] The phrase "systemic administration" as used herein refers to its meaning as is generally accepted in the art. The term generally refers in vivo systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body. SIR-ONC-00001
[0112] The term "target" as it refers to MET refers to any MET target protein, peptide, or polypeptide, such as encoded by Genbank Accession Nos. shown in Table 10. The term also refers to nucleic acid sequences or target polynucleotide sequence encoding any target protein, peptide, or polypeptide, such as proteins, peptides, or polypeptides encoded by sequences having Genbank Accession Nos. shown in Table 10. The target of interest can include target polynucleotide sequences, such as target DNA or target RNA. The term "target" is also meant to include other sequences, such as differing isoforms, mutant target genes, splice variants of target polynucleotides, target polymorphisms, and non-coding (e.g., ncRNA, miRNA, stRNA, sRNA) or other regulatory polynucleotide sequences as described herein.
[0113] The phrase "target site" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to a sequence within a target nucleic acid molecule, (e.g., RNA) that is "targeted", e.g., for cleavage mediated by an siNA construct, which contains sequences within its antisense region that are complementary to the target sequence.
[0114] The phrase "therapeutically effective amount" as used herein refers to its meaning as is generally accepted in the art. The term generally refers to the amount of the compound or composition that will elicit the biological or medical response of a cell, tissue, system, animal or human that is be sought by the researcher, veterinarian, medical doctor or other clinician. For example, if a given clinical treatment is considered effective when there is at least a 25% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is that amount necessary to effect at least a 25% reduction in that parameter.
[0115] The phrase "universal base" as used herein refers to its meaning as is generally accepted in the art. The term universal base generally refers to nucleotide base analogs that form base pairs with each of the natural DNA/RNA bases with little or no discrimination between them. Non-limiting examples of universal bases include C-phenyl, C-naphthyl and other aromatic derivatives, inosine, azole carboxamides, and nitroazole derivatives such as 3- nitropyrrole, 4-nitroindole, 5-nitroindole, and 6-nitroindole as known in the art (see for example, Loakes, 2001, Nucleic Acids Research, 29, 2437-2447). SIR-ONC-00001
[0116] The term "up-regulate" as used herein refers to its meaning as is generally accepted in the art. With reference to exemplary nucleic acid molecules of the invention, the term refers to an increase in the expression of a gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or activity of one or more RNAs, proteins or protein subunits, above that observed in the absence of the nucleic acid molecules (e.g. , siNA) of the invention. In certain instances, up-regulation or promotion of gene expression with an siNA molecule is above that level observed in the presence of an inactive or attenuated molecule. In other instances, up-regulation or promotion of gene expression with siNA molecules is above that level observed in the presence of, for example, an siNA molecule with scrambled sequence or with mismatches. In still other instances, up-regulation or promotion of gene expression with a nucleic acid molecule of the instant invention is greater in the presence of the nucleic acid molecule than in its absence. In some instances, up-regulation or promotion of gene expression is associated with inhibition of RNA mediated gene silencing, such as RNAi mediated cleavage or silencing of a coding or non-coding RNA target that down regulates, inhibits, or silences the expression of the gene of interest to be up-regulated. The down regulation of gene expression can, for example, be induced by a coding RNA or its encoded protein, such as through negative feedback or antagonistic effects. The down regulation of gene expression can, for example, be induced by a non-coding RNA having regulatory control over a gene of interest, for example by silencing expression of the gene via translational inhibition, chromatin structure, methylation, RISC mediated RNA cleavage, or translational inhibition. As such, inhibition or down regulation of targets that down regulate, suppress, or silence a gene of interest can be used to up- regulate expression of the gene of interest toward therapeutic use.
[0117] The term "vector" as used herein refers to its meaning as is generally accepted in the art. The term vector generally refers to any nucleic acid- and/or viral-based expression system or technique used to deliver one or more nucleic acid molecules.
B. siNA Molecules of the Invention
[0118] The present invention provides compositions and methods comprising siNAs targeted to MET that can be used to treat diseases, e.g., malignancies and/or cancers associated with MET expression. In particular aspects and embodiments of the invention, the nucleic acid molecules SIR-ONC-00001 of the invention comprise at least a 15 nucleotide sequence of the sequences shown in Table la and Table lb. The siNAs can be provided in several forms. For example, the siNA can be isolated as one or more siNA compounds, or it may be in the form of a transcriptional cassette in a DNA plasmid. The siNA may also be chemically synthesized and can include modifications as shown, for example, but not limitation, in Table lc and Table 11. The siNAs can be administered alone or co-administered with other siNA molecules or with conventional agents that treat a MET related disease or condition.
[0119] The siNA molecules of the invention can be used to mediate gene silencing, specifically MET, via interaction with RNA transcripts or alternately by interaction with particular gene sequences, wherein such interaction results in modulation of gene silencing either at the transcriptional level or post-transcriptional level such as, for example, but not limited to, RNAi or through cellular processes that modulate the chromatin structure or methylation patterns of the target and prevent transcription of the target gene, with the nucleotide sequence of the target thereby mediating silencing. More specifically, the target is any of MET RNA, DNA, or mRNA,
[0120] In one aspect, the invention provides short interfering nucleic acid (siNA) molecules for inhibiting the expression of the MET gene in a cell or mammal. The siNA can be single- stranded or double- stranded. When double- stranded, the siNA comprising a sense and an antisense stand. The antisense strand is complementary to at least a part of an mRNA formed in the expression of the MET gene. The sense strand comprises a region that is complementary to the antisense strand. In specific embodiments, the antisense strand comprises at least a 15 nucleotide sequence of an antisense sequence listed in Table lb. Generally, the double- stranded siNA comprises at least a 15 nucleotide sequence of the sense strand in Table lb and at least a 15 nucleotide sequence of the antisense strand in Table lb. One or more of the nucleotides of the siNAs of the invention are optionally modified. In further embodiments having modifications, some siNAs of the invention comprises at least one nucleotide sequence selected from the groups of sequences provide in Table lc. In other embodiments, the siNA comprises at least two sequences selected from the group of sequences provided in Table lc, wherein one of the at least two sequences is complementary to another of the at least two sequences and one of the at least two sequences is complementary to a sequence of a mRNA generated in the SIR-ONC-00001 expression of the MET gene. Examples of certain modified siNAs of the invention are in Table lc.
[0121] The double stranded RNA molecules of the invention can comprise two distinct and separate strands that can be symmetric or asymmetric and are complementary, i.e., two single- stranded RNA molecules, or can comprise one single- stranded molecule in which two complementary portions, e.g., a sense region and an antisense region, are base-paired, and are covalently linked by one or more single- stranded "hairpin" areas {i.e. loops) resulting in, for example, a single-stranded short-hairpin polynucleotide or a circular single-stranded polynucleotide.
[0122] The linker can be polynucleotide linker or a non-nucleotide linker. In some embodiments, the linker is a non-nucleotide linker. In some embodiments, a hairpin or circular siNA molecule of the invention contains one or more loop motifs, wherein at least one of the loop portions of the siNA molecule is biodegradable. For example, a single-stranded hairpin siNA molecule of the invention is designed such that degradation of the loop portion of the siNA molecule in vivo can generate a double- stranded siNA molecule with 3'-terminal overhangs, such as 3'-terminal nucleotide overhangs comprising 1, 2, 3 or 4 nucleotides. Or alternatively, a circular siNA molecule of the invention is designed such that degradation of the loop portions of the siNA molecule in vivo can generate a double-stranded siNA molecule with 3'-terminal overhangs, such as 3 '-terminal nucleotide overhangs comprising about 2 nucleotides.
[0123] In symmetric siNA molecules of the invention, each strand, the sense (passenger) strand and antisense (guide) strand, are independently about 15 to about 30 {e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length. Generally, each strand of the symmetric siNA molecules of the invention are about 19-24 {e.g., about 19, 20, 21, 22, 23 or 24) nucleotides in length.
[0124] In asymmetric siNA molecules, the antisense region or strand of the molecule is about 15 to about 30 {e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length, wherein the sense region is about 3 to about 25 {e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides in length. SIR-ONC-00001
Generally, each strand of the asymmetric siNA molecules of the invention is about 19-24 (e.g., about 19, 20, 21, 22, 23 or 24) nucleotides in length.
[0125] In yet other embodiments, siNA molecules of the invention comprise single stranded hairpin siNA molecules, wherein the siNA molecules are about 25 to about 70 (e.g. , about 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 40, 45, 50, 55, 60, 65, or 70) nucleotides in length.
[0126] In still other embodiments, siNA molecules of the invention comprise single-stranded circular siNA molecules, wherein the siNA molecules are about 38 to about 70 (e.g., about 38, 40, 45, 50, 55, 60, 65, or 70) nucleotides in length.
[0127] In still other embodiments, siNA molecules of the invention comprise single-stranded non-circular siNA molecules, wherein the siNA molecules are independently about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length.
[0128] In various symmetric embodiments, the siNA duplexes of the invention independently comprise about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) base pairs. Generally, the duplex structure of siNAs of the invention is between 15 and 30, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 base pairs in length.
[0129] In yet other embodiments, where the duplex siNA molecules of the invention are asymmetric, the siNA molecules comprise about 3 to 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) base pairs. Generally, the duplex structure of siNAs of the invention is between 15 and 25, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 base pairs in length.
[0130] In still other embodiments, where the siNA molecules of the invention are hairpin or circular structures, the siNA molecules comprise about 15 to about 30 (e.g. , about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) base pairs.
[0131] The sense strand and antisense strand, or the sense region and antisense region, of the siNA molecules of the invention can be complementary. Also, the antisense strand or antisense SIR-ONC-00001 region can be complementary to a nucleotide sequence or a portion thereof of the MET target RNA. The sense strand or sense region of the siNA can comprise a nucleotide sequence of a MET gene or a portion thereof. In certain embodiments, the sense region or sense strand of an siNA molecule of the invention is complementary to that portion of the antisense region or antisense strand of the siNA molecule that is complementary to a MET target polynucleotide sequence, such as for example, but not limited to, those sequences represented by GENBANK Accession Nos. shown in Table 10.
[0132] In some embodiments, siNA molecules of the invention have perfect complementarity between the sense strand or sense region and the antisense strand or antisense region of the siNA molecule. In other or the same embodiments, the antisense strand of the siNA molecules of the invention are perfectly complementary to a corresponding target nucleic acid molecule.
[0133] In yet other embodiments, siNA molecules of the invention have partial complementarity (i.e., less than 100% complementarity) between the sense strand or sense region and the antisense strand or antisense region of the siNA molecule or between the antisense strand or antisense region of the siNA molecule and a corresponding target nucleic acid molecule. Thus, in some embodiments, the double- stranded nucleic acid molecules of the invention, have between about 15 to about 30 (e.g. , about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in one strand that are complementary to the nucleotides of the other strand. In other embodiments, the molecules have between about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in the sense region that are complementary to the nucleotides of the antisense region, of the double-stranded nucleic acid molecule. In certain embodiments, the double- stranded nucleic acid molecules of the invention have between about 15 to about 30 (e.g. , about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in the antisense strand that are complementary to a nucleotide sequence of its corresponding target nucleic acid molecule.
[0134] In other embodiments, the siNA molecule can contain one or more nucleotide deletions, substitutions, mismatches and/or additions; provided, however, that the siNA molecule maintains its activity, for example, to mediate RNAi. In a non-limiting example, the deletion, SIR-ONC-00001 substitution, mismatch and/or addition can result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair. Thus, in some embodiments, for example, the double-stranded nucleic acid molecules of the invention, have 1 or more (e.g., 1, 2, 3, 4, 5, or 6) nucleotides, in one strand or region that are mismatches or non-base-paired with the other strand or region.. In other embodiments, the double- stranded nucleic acid molecules of the invention, have 1 or more (e.g., 1, 2, 3, 4, 5, or 6) nucleotides in each strand or region that are mismatches or non-base-paired with the other strand or region. In a preferred embodiment, the siNA of the invention contains no more than 3 mismatches. If the antisense strand of the siNA contains mismatches to a target sequence, it is preferable that the area of mismatch not be located in the center of the region of complementarity.
[0135] In other embodiments, the siNA molecule can contain one or more nucleotide deletions, substitutions, mismatches and/or additions to a sequence in Table lb provided, however, that the siNA molecule maintains its activity, for example, to mediate RNAi. In a non- limiting example, the deletion, substitution, mismatch and/or addition can result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair.
[0136] The invention also comprises double- stranded nucleic acid (siNA) molecules as otherwise described hereinabove in which the first strand and second strand are complementary to each other and wherein at least one strand is hybridisable to the polynucleotide sequence of a sequence in Table lb under conditions of high stringency, and wherein any of the nucleotides is unmodified or chemically modified.
[0137] Hybridization techniques are well known to the skilled artisan (see for instance, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)). Preferred stringent hybridization conditions include overnight incubation at 42°C in a solution comprising: 50% formamide, 5xSSC (150mM NaCl, 15mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution, 10% dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters in O. lx SSC at about 65°C.
[0138] In one specific embodiment, the first strand has about 15, 16, 17, 18, 19, 20 or 21 nucleotides that are complementary to the nucleotides of the other strand and at least one strand SIR-ONC-00001 is hybridisable to a polynucleotide sequence in Table lb. In a more preferred embodiment, the first strand has about 15, 16, 17, 18, 19, 20 or 21 nucleotides that are complementary to the nucleotides of the other strand and at least one strand is hybridisable to SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; under conditions of high stringency, and wherein any of the nucleotides is unmodified or chemically modified.
[0139] In certain embodiments, the siNA molecules of the invention comprise overhangs of about 1 to about 4 (e.g. , about 1, 2, 3 or 4) nucleotides. The nucleotides in the overhangs can be the same or different nucleotides. In some embodiments, the overhangs occur at the 3 '-end at one or both strands of the double-stranded nucleic acid molecule. For example, a double- stranded nucleic acid molecule of the invention can comprise a nucleotide or non-nucleotide overhang at the 3 '-end of the antisense strand/region, the 3 '-end of the sense strand/region, or both the antisense strand/region and the sense strand/region of the double-stranded nucleic acid molecule.
[0140] In some embodiments, the nucleotides comprising the overhang portion of an siNA molecule of the invention comprise sequences based on the MET target polynucleotide sequence in which nucleotides comprising the overhang portion of the antisense strand/region of an siNA molecule of the invention can be complementary to nucleotides in the MET target polynucleotide sequence and/or nucleotides comprising the overhang portion of the sense strand/region of an siNA molecule of the invention can comprise the nucleotides in the MET target polynucleotide sequence. Thus, in some embodiments, the overhang comprises a two nucleotide overhang that is complementary to a portion of the MET target polynucleotide sequence. In other embodiments, however, the overhang comprises a two nucleotide overhang that is not complementary to a portion of the MET target polynucleotide sequence. In certain embodiments, the overhang comprises a 3'-UU overhang that is not complementary to a portion of the MET target polynucleotide sequence. In other embodiments, the overhang comprises a UU overhang at the 3' end of the antisense strand and a TT overhang at the 3' end of the sense strand. In other embodiments, the overhang comprises nucleotides as described in the examples, Tables, and Figures herein. SIR-ONC-00001
[0141] In any of the embodiments of the siNA molecules described herein having 3'-terminal nucleotide overhangs, the overhangs are optionally chemically modified at one or more nucleic acid sugar, base, or backbone positions. Representative, but not limiting examples of modified nucleotides in the overhang portion of a double- stranded nucleic acid (siNA) molecule of the invention include: 2'-0-alkyl (e.g., 2'-0-methyl), 2'-deoxy, 2'-deoxy-2'-fluoro, 2'-deoxy-2'- fluoroarabino (FANA), 4'-thio, 2'-0-trifluoromethyl, 2'-0-ethyl-trifluoromethoxy, 2'-0- difluoromethoxy-ethoxy, universal base, acyclic, or 5-C-methyl nucleotides. In more preferred embodiments, the overhang nucleotides are each independently, a 2'-0-alkyl nucleotide, a 2'-0- methyl nucleotide, a 2'-dexoy-2-fluoro nucleotide, or a 2'-deoxy ribonucleotide. In some instances the overhang nucleotides are linked by a one or more phosphorothioate linkages.
[0142] In yet other embodiments, siNA molecules of the invention comprise duplex nucleic acid molecules with blunt ends (i.e., without nucleotide overhangs), where both ends are blunt, or alternatively, where one of the ends is blunt. In some embodiments, the siNA molecules of the invention can comprises one blunt end, for example wherein the 5 '-end of the antisense strand and the 3 '-end of the sense strand do not have any overhanging nucleotides. In another example, the siNA molecule comprises one blunt end, for example wherein the 3 '-end of the antisense strand and the 5 '-end of the sense strand do not have any overhanging nucleotides. In other embodiments, siNA molecules of the invention comprise two blunt ends, for example wherein the 3 '-end of the antisense strand and the 5 '-end of the sense strand as well as the 5 '-end of the antisense strand and 3 '-end of the sense strand do not have any overhanging nucleotides.
[0143] In any of the embodiments or aspects of the siNA molecules of the invention, the sense strand and/or the antisense strand can further have a cap, such as described herein or as known in the art, at the 3'-end, the 5'-end, or both of the 3' and 5'-ends of the sense strand and/or antisense strand. Or as in the case of a hairpin siNA molecule, the cap can be at either one or both of the terminal nucleotides of the polynucleotide. In some embodiments, the cap is at one of both of the ends of the sense strand of a double- stranded siNA molecule. In other embodiments, the cap is at the 3 '-end of antisense (guide) strand. In preferred embodiments, the caps are at the 3'-end of the sense strand and the 5'-end of the sense strand. SIR-ONC-00001
[0144] Representative, but non-limiting examples of such terminal caps include an inverted abasic nucleotide, an inverted deoxy abasic nucleotide, an inverted nucleotide moiety, a group shown in Figure 5, a glyceryl modification, an alkyl or cycloalkyl group, a heterocycle, or any other cap as is generally known in the art.
[0145] Any of the embodiments of the siNA molecules of the invention can have a 5' phosphate termini. In some embodiments, the siNA molecules lack terminal phosphates.
[0146] Any siNA molecule or construct of the invention can comprise one or more chemical modifications. Modifications can be used to improve in vitro or in vivo characteristics such as stability, activity, toxicity, immune response (e.g., prevent stimulation of an interferon response, an inflammatory or pro -inflammatory cytokine response, or a Toll-like Receptor (TIF) response), and/or bioavailability.
[0147] Applicants describe herein chemically modified siNA molecules with improved RNAi activity and/or stability compared to corresponding unmodified siNA molecules. Various chemically modified siNA motifs disclosed herein provide the capacity to maintain RNAi activity that is substantially similar to unmodified or minimally modified active siRNA (see for example Elbashir et al., 2001, EMBO J., 20:6877-6888) while at the same time providing nuclease resistance and pharmacokinetic properties suitable for use in therapeutic applications.
[0148] In various embodiments, the siNA molecules of the invention comprise modifications wherein any (e.g., one or more or all) nucleotides present in the sense and/or antisense strand are modified nucleotides (e.g., wherein one nucleotide is modified, some nucleotides (i.e., plurality or more than one) are modified, or all nucleotides are modified nucleotides. In some embodiments, the siNA molecules of the invention are partially modified (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 55, or 59 nucleotides are modified) with chemical modifications. In some embodiments, an siNA molecule of the invention comprises at least about 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, or 60 nucleotides that are modified nucleotides. In other embodiments, the siNA molecules of the invention are completely modified (e.g., 100% modified) with chemical modifications, i.e., the siNA molecule does not contain any SIR-ONC-00001 ribonucleotides. In some of embodiments, one or more of the nucleotides in the sense strand of the siNA molecules of the invention are modified. In the same or other embodiments, one or more of the nucleotides in the antisense strand of the siNA molecules of the invention are modified.
[0149] The chemical modification within a single siNA molecule can be the same or different. In some embodiments, at least one strand has at least one chemical modification. In other embodiments, each strand has at least one chemical modifications, which can be the same or different, such as, sugar, base, or backbone (i.e., internucleotide linkage) modifications. In other embodiments, siNA molecules of the invention contain at least 2, 3, 4, 5, or more different chemical modifications.
[0150] Non-limiting examples of chemical modifications that are suitable for use in the present invention, are disclosed in U.S. Patent Applications Nos. 10/444,853; 10/981,966; 12/064,014 and in references cited therein and include sugar, base, and phosphate, non- nucleotide modifications, and/or any combination thereof.
[0151] In certain specific embodiments of the invention, at least one modified nucleotide is a 2'-deoxy-2-fluoro nucleotide, a 2'-deoxy nucleotide, a 2'-0-alkyl (e.g., 2'-0-methyl) nucleotide, or a locked nucleic acid (LNA) nucleotide as is generally recognized in the art.
[0152] In yet other embodiment of the invention, at least one nucleotide has a ribo-like, Northern or A form helix configuration (see e.g., Saenger, Principles of Nucleic Acid Structure, Springer- Verlag ed., 1984). Non-limiting examples of nucleotides having a Northern configuration include locked nucleic acid (LNA) nucleotides (e.g., 2'-0, 4'-C-methylene-(D- ribofuranosyl) nucleotides); 2'-methoxyethoxy (MOE) nucleotides; 2'-methyl-thio-ethyl nucleotides, 2'-deoxy-2'-fluoro nucleotides; 2'-deoxy-2'-chloro nucleotides; 2'-azido nucleotides; 2'-0-trifluoromethyl nucleotides; 2'-0-ethyl-trifluoromethoxy nucleotides; 2'-0- difluoromethoxy-ethoxy nucleotides; 4'-thio nucleotides and 2'-0-methyl nucleotides.
[0153] In various embodiments, a majority (e.g., greater than 50%) of the pyrimidine nucleotides present in the double- stranded siNA molecule comprises a sugar modification. In some of the same and/or other embodiments, a majority (e.g., greater than 50%) of the purine nucleotides present in the double-stranded siNA molecule comprises a sugar modification. SIR-ONC-00001
[0154] In some embodiments, the pyrimidine nucleotides in the antisense strand are 2'-0- methyl or 2'-deoxy-2'-fluoro pyrimidine nucleotides and the purine nucleotides present in the antisense strand are 2'-0-methyl nucleotides or 2'-deoxy nucleotides. In other embodiments, the pyrimidine nucleotides in the sense strand are 2'-deoxy-2'-fluoro pyrimidine nucleotides and the purine nucleotides present in the sense strand are 2'-0-methyl or 2'-deoxy purine nucleotides.
[0155] In certain embodiments of the invention, all the pyrimidine nucleotides in the complementary region on the sense strand are 2'-deoxy-2'-fluoro pyrimidine nucleotides. In certain embodiments, all of the pyrimidine nucleotides in the complementary region of the antisense strand are 2'-deoxy-2'-fluoro pyrimidine nucleotides. In certain embodiments, all the purine nucleotides in the complementary region on the sense strand are 2'-deoxy purine nucleotides. In certain embodiments, all of the purines in the complementary region on the antisense strand are 2'-0-methyl purine nucleotides. In certain embodiments, all of the pyrimidine nucleotides in the complementary regions on the sense strand are 2'-deoxy-2'-fluoro pyrimidine nucleotides; all of the pyrimidine nucleotides in the complementary region of the antisense strand are 2'-deoxy-2'-fluoro pyrimidine nucleotides; all the purine nucleotides in the complementary region on the sense strand are 2'-deoxy purine nucleotides and all of the purines in the complementary region on the antisense strand are 2'-0-methyl purine nucleotides.
[0156] In some embodiments, at least 5 or more of the pyrimidine nucleotides in one or both stands are 2' -deoxy-2' -fluoro pyrimidine nucleotides. In some embodiments, at least 5 or more of the pyrimidine nucleotides in one or both stands are 2'-0-methyl pyrimidine nucleotides. In some embodiments, at least 5 or more of the purine nucleotides in one or both stands are 2'- deoxy-2' -fluoro purine nucleotides In some embodiments, at least 5 or more of the purine nucleotides in one or both stands are 2'-0-methyl purine nucleotides.
[0157] In certain embodiments, the purines and pyrimidines are differentially modified at the 2'-sugar position (i.e., at least one purine has a different modification from at least one pyrimidine in the same or different strand at the 2'-sugar position). For example, in some instances, at least 5 or more of the pyrimidine nucleotides in one or both stands are 2' -deoxy-2' - fluoro pyrimidine nucleotides and at least 5 or more purine nucleotides in one or both strands are 2'-0-methyl purine nucleotides. In other instances at least 5 or more of the pyrimidine SIR-ONC-00001 nucleotides in one or both stands are 2'-0-methyl pyrimidine nucleotides and at least 5 or more purine nucleotides in one or both strands are 2'-deoxy-2'-fluoro purine nucleotides.
[0158] Further non-limiting examples of sense and antisense strands of such siNA molecules having various modifications and modifications patterns are shown in Figures 2 and 3.
[0159] Any of the above described modifications, or combinations thereof, including those in the references cited, can be applied to any of the siNA molecules of the invention.
[0160] The modified siNA molecules of the invention can comprise modifications at various locations within the siNA molecule. In some embodiments, the double-stranded siNA molecule of the invention comprises modified nucleotides at internal base paired positions within the siNA duplex. In other embodiments, a double- stranded siNA molecule of the invention comprises modified nucleotides at non-base paired or overhang regions of the siNA molecule. In yet other embodiments, a double- stranded siNA molecule of the invention comprises modified nucleotides at terminal positions of the siNA molecule. For example, such terminal regions include the 3'- position and/or 5 '-position of the sense and/or antisense strand or region of the siNA molecule. Additionally, any of the modified siNA molecules of the invention can have a modification in one or both oligonucleotide strands of the siNA duplex, for example in the sense strand, the antisense strand, or both strands. Moreover, with regard to chemical modifications of the siNA molecules of the invention, each strand of the double-stranded siNA molecules of the invention can have one or more chemical modifications, such that each strand comprises a different pattern of chemical modifications.
[0161] In certain embodiments each strand of a double-stranded siNA molecule of the invention comprises a different pattern of chemical modifications, such as any Stab modification chemistries described herein (see Table 11) or any combination thereof, i.e., different combinations of defined Stabilzation chemistry (Stab) sense and antisense strands. Further, non- limiting examples of modification schemes that could give rise to different patterns of modifications are shown in Table 11. The stabilization chemistries referred to in Table 11 as Stab, can be combined in any combination of sense/antisense chemistries, such as Stab 7/8, Stab 7/11, Stab 8/8, Stab 18/8, Stab 18/11, Stab 12/13, Stab 7/13, Stab 18/13, Stab 7/19, Stab 8/19, SIR-ONC-00001
Stab 18/19, Stab 7/20, Stab 8/20, Stab 18/20, Stab 7/32, Stab 8/32, or Stab 18/32 or any other combination of Stabilization chemistries.
[0162] In any of the siNAs of the invention, one or more (for example 1, 2, 3, 4 or 5) nucleotides at the 5 '-end of the guide strand or guide region (also known as antisense strand or antisense region) of the siNA molecule are ribonucleotides.
[0163] In certain embodiments, the present invention provides a double- stranded short interfering nucleic acid (siNA) molecule that modulates the expression of MET, wherein the siNA comprises a sense strand and an antisense strand; each strand is independently 15 to 30 nucleotides in length; and the antisense strand comprises at least 15 nucleotides having sequence complementary to any of:
5'-GUGGCACUUUACUUACUUU-3' (SEQ ID NO: 1);
5'-UUUACUUACUUUAACUGGA-3' (SEQ ID NO: 43);
5'-CAAGCAUCUUCAGUUACCG-3' (SEQ ID NO: 51); or
5 ' - AGAAGGAAGUGUUU AAUAU-3' (SEQ ID NO: 53).
[0164] In some embodiments, the antisense strand of a siNA molecule of the invention comprises at least a 15 nucleotide sequence of:
5 ' - AAAGUAAGUAAAGUGCC AC-3' (SEQ ID NO: 1049);
5'-UCCAGUUAAAGUAAGUAAA-3' (SEQ ID NO: 1091);
5'-CGGUAACUGAAGAUGCUUG-3' (SEQ ID NO: 1099); or
5'-AUAUUAAACACUUCCUUCU-3' (SEQ ID NO: 1101).
[0165] In some embodiments, the sense strand of a siNA molecule of the invention comprises at least a 15 nucleotide sequence of:
5'-GUGGCACUUUACUUACUUU-3' (SEQ ID NO: 1); 5'-UUUACUUACUUUAACUGGA-3' (SEQ ID NO: 43);
5'-CAAGCAUCUUCAGUUACCG-3' (SEQ ID NO: 51); or SIR-ONC-00001
5 ' - AGAAGGAAGUGUUU AAUAU-3' (SEQ ID NO: 53).
[0166] In some embodiments, a siNA molecule of the invention comprises any of:
5'-GUGGCACUUUACUUACUUU-3' (SEQ ID NO: 1) and 5'- AAAGUAAGUAAAGUGCCAC-3' (SEQ ID NO: 1049); or
5'-UUUACUUACUUUAACUGGA-3' (SEQ ID NO: 43) and 5'- UCCAGUUAAAGUAAGUAAA-3' (SEQ ID NO: 1091); or
5'-CAAGCAUCUUCAGUUACCG-3' (SEQ ID NO: 51) and 5'- CGGUAACUGAAGAUGCUUG-3' (SEQ ID NO: 1099); or
5' - AGAAGGAAGUGUUU AAUAU-31 (SEQ ID NO: 53) and 5'- AUAUUAAACACUUCCUUCU-31 (SEQ ID NO: 1101).
[0167] Any of the above described modifications, or combinations thereof, including those in the references cited, can be applied to any of these embodiments.
[0168] In certain embodiments, the nucleotides of the at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101 form a contiguous stretch of nucleotides.
[0169] In some embodiments, the siNA molecule can contain one or more nucleotide deletions, substitutions, mismatches and/or additions to the at least 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; provided, however, that the siNA molecule maintains its activity, for example, to mediate RNAi. In a non-limiting example, the deletion, substitution, mismatch and/or addition can result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair.
[0170] In certain embodiments of the invention, double- stranded siNA molecules are provided, wherein the molecule has a sense strand and an antisense strand and comprises the following formula (A): SIR-ONC-00001
B NX3 — (Ν)χ2 B -3
B (N)xi NX4 [Ν]χ5 -5'
(A) wherein, the upper strand is the sense strand and the lower strand is the antisense strand of the double- stranded nucleic acid molecule; wherein the antisense strand comprises at least a 15 nucleotide sequence of SEQ ID NO: 1049, SEQ ID NO: 1091, SEQ ID NO: 1099, or SEQ ID NO: 1101, and the sense strand comprises a sequence having complementarity to the antisense strand; each N is independently a nucleotide which is unmodified or chemically modified or a non-nucleotide; each B is a terminal cap that is present or absent;
(N) represents overhanging nucleotides, each of which is independently unmodified or chemically modified;
[N] represents nucleotides that are ribonucleotides; XI and X2 are independently integers from 0 to 4; X3 is an integer from 15 to 30; X4 is an integer from 9 to 30; and
X5 is an integer from 0 to 6, provided that the sum of X4 and X5 is 15-30.
[0171] In certain embodiments, the nucleotides of the at least a 15 nucleotide sequence of SEQ ID NO: 1049, SEQ ID NO: 1091, SEQ ID NO: 1099, or SEQ ID NO: 1101 form a contiguous stretch of nucleotides.
[0172] In some embodiments, the siNA molecule of formula A can contain one or more nucleotide deletions, substitutions, mismatches and/or additions to the at least 15 nucleotide sequence of SEQ ID NO: 1049, SEQ ID NO: 1091, SEQ ID NO: 1099, or SEQ ID NO: 1101; provided, however, that the siNA molecule maintains its activity, for example, to mediate RNAi. SIR-ONC-00001
In a non-limiting example, the deletion, substitution, mismatch and/or addition can result in a loop or bulge, or alternately a wobble or other alternative (non Watson-Crick) base pair.
[0173] In one embodiment, the invention features a double- stranded short interfering nucleic acid (siNA) of formula (A); wherein
(a) one or more pyrimidine nucleotides in Νχ4 positions are independently 2'-deoxy- 2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof;
(b) one or more purine nucleotides in Νχ4 positions are independently 2'-deoxy-2'- fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof;
(c) one or more pyrimidine nucleotides in Νχ3 positions are independently 2'-deoxy- 2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof; and
(d) one or more purine nucleotides in Νχ3 positions are independently 2'-deoxy-2'- fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof.
[0174] In certain embodiments, the invention features a double-stranded short interfering nucleic acid (siNA) molecule of formula (A); wherein
(a) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ4 positions are 2'-deoxy-2'- fluoro nucleotides;
(b) 1, 2, 3, 4, 5 or more purine nucleotides in Νχ4 positions are 2'-0-alkyl nucleotides;
(c) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ3 positions are 2'-deoxy-2'- fluoro nucleotides; and
(d) 1, 2, 3, 4, 5 or more purine nucleotides in Nxs positions are 2'-deoxy nucleotides.
[0175] In certain embodiments, the invention features a double-stranded short interfering nucleic acid (siNA) molecule of formula (A); wherein SIR-ONC-00001
(a) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ4 positions are 2'-0-alkyl nucleotides;
(b) 1, 2, 3, 4, 5 or more purine nucleotides in Νχ4 positions are ribonucleotides;
(c) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ3 positions are 2'-0-alkyl nucleotides; and
(d) 1, 2, 3, 4, 5 or more purine nucleotides in positions are ribonucleotides.
[0176] In certain embodiments, the invention features a double-stranded short interfering nucleic acid (siNA) molecule of formula (A); wherein
(a) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ4 positions are 2'-deoxy-2'- fluoro nucleotides;
(b) 1, 2, 3, 4, 5 or more purine nucleotides in Νχ4 positions are 2'-0-alkyl nucleotides;
(c) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ3 positions are 2'-0-alkyl nucleotides; and
(d) 1, 2, 3, 4, 5 or more purine nucleotides in Νχ3 positions are 2'-deoxy-2'-fluoro nucleotides.
[0177] In certain embodiments, the invention features a double-stranded short interfering nucleic acid (siNA) molecule of formula (A) further comprising one or more phosphorothioate internucleotide linkages.
[0178] In some embodiments, siNA molecules having formula A comprise a terminal phosphate group at the 5 '-end of the antisense strand or antisense region of the nucleic acid molecule.
[0179] In various embodiments, siNA molecules having formula A comprise X5 = 0, 1, 2, or 3; each XI and X2 = 1 or 2; X3 = 18, 19, 20, 21, 22, or 23, and X4 = 17, 18, 19, 20, 21, 22, or 23.
[0180] In certain embodiments, siNA molecules having formula A comprise X5 = 3. In other embodiments siNA molecules having formula A comprise X5 = 0. SIR-ONC-00001
[0181] In certain embodiments, siNA molecules having formula A comprise XI = 2 and X2 = 2.
[0182] In various embodiments, siNA molecules having formula A comprise X5 = 0, XI = 2, and X2 = 2. In other embodiments, siNA molecules having formula A comprise X5 = 3, XI = 2, and X2 = 2.
[0183] In one specific embodiment, an siNA molecule having formula A comprises X5 = 3; each XI and X2 = 2; X3 = 19, and X4 = 16..
[0184] In another specific embodiment, an siNA molecule having formula A comprises X5 = 0; each XI and X2 = 2; X3 = 19, and X4 = 19.
[0185] In certain embodiments, siNA molecules having formula A comprise caps (B) at the 3' and 5' ends of the sense strand or sense region.
[0186] In certain embodiments, siNA molecules having formula A comprise caps (B) at the 3 '-end of the antisense strand or antisense region.
[0187] In various embodiments, siNA molecules having formula A comprise caps (B) at the 3' and 5' ends of the sense strand or sense region and caps (B) at the 3 '-end of the antisense strand or antisense region.
[0188] In yet other embodiments, siNA molecules having formula A comprise caps (B) only at the 5'-end of the sense (upper) strand of the double- stranded nucleic acid molecule.
[0189] In some embodiments, siNA molecules having formula A further comprise one or more phosphorothioate intemucleotide linkages between the nucleotides. In certain embodiments, siNA molecules having formula A comprise one or more phosphorothioate intemucleotide linkages between the first terminal (N) and the adjacent nucleotide on the 3 'end of the sense strand, antisense strand, or both sense strand and antisense strands of the nucleic acid molecule. For example, a double-stranded nucleic acid molecule can comprise XI and/or X2 = 2 having overhanging nucleotide positions with a phosphorothioate intemucleotide linkage, e.g., (NsN) where "s" indicates phosphorothioate. SIR-ONC-00001
[0190] In some embodiments, one or more of the nucleotides of siNA molecules having formula A have a universal base.
[0191] In certain embodiments, siNA molecules having formula A have at position 14 from the 5'-end of the antisense strand a ribonucleotide when the nucleotide at that position 14 is a purine. In other embodiments, siNA molecules having formula A have at position 14 from the 5'-end of the antisense strand a ribonucleotide, a 2'-deoxy-2'-fluoro nucleotide or a 2'-0-methyl nucleotide when the nucleotide at that position 14 is a pyrimidine nucleotide.
[0192] In some embodiments, siNA molecules having formula A comprises (N) nucleotides in the antisense strand (lower strand) that are complementary to nucleotides in a MET target polynucleotide sequence, which also has complementarity to the N and [N] nucleotides of the antisense (lower) strand.
[0193] Any of the above described modifications, or combinations thereof, discussed above as applicable to siNAs of the invention, including those in the references cited, can be applied to any of the embodiments to siNA molecules having formula A.
C. Generation/Synthesis of siNA Molecules
[0194] The siNAs of the invention can be obtained using a number of techniques known to those of skill in the art. For example the siNA can be chemically synthesized or may be encoded by plasmid (e.g., transcribed as sequences that automatically fold into duplexes with hairpin loops.). siNA can also be generated by cleavage of longer dsRNA (e.g., dsRNA greater than about 25 nucleotides in length) by the E coli RNase II or Dicer. These enzymes process the dsRNA into biologically active siNA (see, e.g., Yang et al, PNAS USA 99:9942-9947 (2002); Calegari et al. PNAS USA 99: 14236 (2002) Byron et al. Ambion Tech Notes; 10 (l):4-6 (2009); Kawaski et al., Nucleic Acids Res., 31:981-987 (2003), Knight and Bass, Science, 293:2269- 2271 (2001) and Roberston et al, J. Biol. Chem 243:82(1969).
1. Chemical Synthesis
[0195] Preferably, siNA of the invention are chemically synthesized. Oligonucleotides (e.g., certain modified oligonucleotides or portions of oligonucleotides lacking ribonucleotides) are SIR-ONC-00001 synthesized using protocols known in the art, for example as described in Caruthers et al, 1992, Methods in Enzymology 211, 3-19, Thompson et al., International PCT Publication No. WO 99/54459, Wincott et al, 1995, Nucleic Acids Res. 23, 2677-2684, Wincott et al, 1997, Methods Mol. Bio., 74, 59, Brennan et al, 1998, Biotechnol Bioeng., 61, 33-45, and Brennan, U.S. Pat. No. 6,001,311. The synthesis of oligonucleotides makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end.
[0196] siNA molecules without modifications are synthesized using procedures as described in Usman et al, 1987, J. Am. Chem. Soc, 109, 7845; Scaringe et al, 1990, Nucleic Acids Res., 18, 5433. These syntheses makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end that can be used for certain siNA molecules of the invention.
[0197] In certain embodiments, the siNA molecules of the invention are synthesized, deprotected, and analyzed according to methods described in U.S. Patent Nos. 6,995,259, 6,686,463, 6,673,918, 6,649,751, 6,989,442, and U.S. Patent Application No. 10/190,359.
[0198] In a non-limiting synthesis example, small scale syntheses are conducted on a 394 Applied Biosystems, Inc. synthesizer using a 0.2 μιηοΐ scale protocol with a 2.5 min coupling step for 2'-0-methylated nucleotides and a 45 second coupling step for 2'-deoxy nucleotides or 2'-deoxy-2'-fluoro nucleotides. Table 12 outlines the amounts and the contact times of the reagents used in the synthesis cycle.
[0199] Alternatively, the siNA molecules of the present invention can be synthesized separately and joined together post-synthetically, for example, by ligation (Moore et al, 1992, Science 256, 9923; Draper et al, International PCT Publication No. WO 93/23569; Shabarova et al, 1991, Nucleic Acids Research 19, 4247; Bellon et al, 1997, Nucleosides & Nucleotides, 16, 951; Bellon et al, 1997, Bioconjugate Chem. 8, 204), or by hybridization following synthesis and/or deprotection.
[0200] Various siNA molecules of the invention can also be synthesized using the teachings of Scaringe et al, US Patent Nos. 5,889,136; 6,008,400; and 6,111,086.
2. Vector Expression SIR-ONC-00001
[0201] Alternatively, siNA molecules of the invention that interact with and down-regulate gene encoding target MET molecules can be expressed and delivered from transcription units (see for example Couture et al, 1996, TIG., 12, 510) inserted into DNA or RNA vectors. The recombinant vectors can be DNA plasmids or viral vectors. siNA expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus, retrovirus, adenovirus, or alphavirus.
[0202] In some embodiments, pol III based constructs are used to express nucleic acid molecules of the invention. Transcription of the siNA molecule sequences can be driven from a promoter for eukaryotic RNA polymerase I (pol I), RNA polymerase II (pol II), or RNA polymerase III (pol III), (see for example, Thompson, U.S. Patent. Nos. 5,902,880 and 6,146,886). (See also, Izant and Weintraub, 1985, Science, 229, 345; McGarry and Lindquist, 1986, Proc. Natl. Acad. Set, USA 83, 399; Scanlon et al, 1991, Proc. Natl. Acad. Sci. USA, 88, 10591-5; Kashani-Sabet et al., 1992, Antis ens e Res. Dev., 2, 3-15; Dropulic et al., 1992, J. Virol., 66, 1432-41; Weerasinghe et al, 1991, J. Virol, 65, 5531-4; Ojwang et al, 1992, Proc. Natl. Acad. Sci. USA, 89, 10802-6; Chen et al., 1992, Nucleic Acids Res., 20, 4581-9; Sarver et al., 1990 Science, 247, 1222-1225; Thompson et al, 1995, Nucleic Acids Res., 23, 2259; Good et al, 1997, Gene Therapy, 4, 45. Transcripts from pol II or pol III promoters are expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type depends on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby. Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Elroy-Stein and Moss, 1990, Proc. Natl. Acad. Sci. U S A, 87, 6743-7; Gao and Huang 1993, Nucleic Acids Res., 21, 2867-72; Lieber et al, 1993, Methods Enzymol, 217, 47-66; Zhou et al, 1990, Mol. Cell. Biol, 10, 4529-37). Several investigators have demonstrated that nucleic acid molecules expressed from such promoters can function in mammalian cells {e.g. Kashani-Sabet et al, 1992, Antisense Res. Dev., 2, 3-15; Ojwang et al, 1992, Proc. Natl. Acad. Sci. U S A, 89, 10802-6; Chen et al, 1992, Nucleic Acids Res., 20, 4581- 9; Yu et al, 1993, Proc. Natl. Acad. Sci. U S A, 90, 6340-4; L'Huillier et al, 1992, EMBO J., 11, 4411-8; Lisziewicz et al, 1993, Proc. Natl. Acad. Sci. U. S. A, 90, 8000-4; Thompson et al, 1995, Nucleic Acids Res., 23, 2259; Sullenger & Cech, 1993, Science, 262, 1566). More specifically, transcription units such as the ones derived from genes encoding U6 small nuclear (snRNA), transfer RNA (tRNA) and adenovirus VA RNA are useful in generating high SIR-ONC-00001 concentrations of desired RNA molecules such as siNA in cells (Thompson et al., supra; Couture and Stinchcomb, 1996, supra; Noonberg et al., 1994, Nucleic Acid Res., 22, 2830; Noonberg et al., U.S. Pat. No. 5,624,803; Good et al., 1997, Gene Ther., 4, 45; Beigelman et al., International PCT Publication No. WO 96/18736. The above siNA transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated virus vectors), or viral RNA vectors (such as retroviral or alphavirus vectors) (for a review see Couture and Stinchcomb, 1996, supra).
[0203] Vectors used to express the siNA molecules of the invention can encode one or both strands of an siNA duplex, or a single self-complementary strand that self hybridizes into an siNA duplex. The nucleic acid sequences encoding the siNA molecules of the instant invention can be operably linked in a manner that allows expression of the siNA molecule (see for example Paul et al., 2002, Nature Biotechnology, 19, 505; Miyagishi and Taira, 2002, Nature Biotechnology, 19, 497; Lee et al, 2002, Nature Biotechnology, 19, 500; and Novina et al, 2002, Nature Medicine, advance online publication doi: 10.1038/nm725).
D. Carrier/Delivery Systems
[0204] The siNA molecules of the invention are added directly, or can be complexed with cationic lipids, packaged within liposomes, or as a recombinant plasmid or viral vectors which express the siNA molecules, or otherwise delivered to target cells or tissues. Methods for the delivery of nucleic acid molecules are described in Akhtar et al., 1992, Trends Cell Bio., 2, 139; Delivery Strategies for Antisense Oligonucleotide Therapeutics, ed. Akhtar, 1995, Maurer et al., 1999, Mol. Membr. Biol., 16, 129-140; Hofland and Huang, 1999, Handb. Exp. Pharmacol., 137, 165-192; and Lee et al., 2000, ACS Symp. Ser., 752, 184-192. Beigelman et al., U.S. Pat. No. 6,395,713 and Sullivan et al., PCT WO 94/02595 further describe the general methods for delivery of nucleic acid molecules. These protocols can be utilized for the delivery of virtually any nucleic acid molecule. Nucleic acid molecules can be administered to cells by a variety of methods known to those of skill in the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as biodegradable polymers, hydrogels, cyclodextrins (see for example, Gonzalez et al., 1999, Bioconjugate Chem., SIR-ONC-00001
10, 1068-1074; Wang et al, International PCT Publication Nos. WO 03/47518 and WO 03/46185), poly(lactic-co-glycolic)acid (PLGA) and PLCA microspheres (see for example US Patent 6,447,796 and US Patent Application Publication No. US 2002130430), biodegradable nanocapsules, and bioadhesive microspheres, or by proteinaceous vectors (O'Hare and Normand, International PCT Publication No. WO 00/53722).
[0205] In one aspect, the present invention provides carrier systems containing the siNA molecules described herein. In some embodiments, the carrier system is a lipid-based carrier system, cationic lipid, or liposome nucleic acid complexes, a liposome, a micelle, a virosome, a lipid nanoparticle or a mixture thereof. In other embodiments, the carrier system is a polymer- based carrier system such as a cationic polymer-nucleic acid complex. In additional embodiments, the carrier system is a cyclodextrin-based carrier system such as a cyclodextrin polymer-nucleic acid complex. In further embodiments, the carrier system is a protein-based carrier system such as a cationic peptide-nucleic acid complex. Preferably, the carrier system in a lipid nanoparticle ("LNP") formulation.
In certain embodiments, the siNA molecules of the invention are formulated with a lipid nanoparticle composition such as is described in U.S. Patent Application Nos. 11/353,630, 11/586,102, 61/189,295, 61/204,878, 61/235,476, 61/249,807, and 61/298,022. In certain preferred embodiments, the siNA molecules of the invention are formulated with a lipid nanoparticle composition comprising a cationic lipid/Cholesterol/PEG-C-DMA/DSPC in a 40/48/2/10 ratio or a cationic lipid/Cholesterol/PEG-DMG/DSPC in a 40/48/2/10 ratio. In more preferred embodiments, the cationic lipid is DLinDMA (see Table 14) and the PEG is PEG- DMG. In a still more preferred embodiments, the N/P ratio of the formulation is 2.8.
[0206] In various embodiments, lipid nanoparticle formulations described in Table 13 are applied to any siNA molecule or combination of siNA molecules herein. In some embodiments, the invention features a composition comprising an siNA molecule of the invention formulated as any of formulation LNP-051; LNP-053; LNP-054; LNP-069; LNP-073; LNP-077; LNP-080; LNP-082; LNP-083; LNP-060; LNP-061; LNP-086; LNP-097; LNP-098; LNP-099; LNP-100; LNP-101; LNP-102; LNP-103; or LNP-104 (see Table 13). SIR-ONC-00001
[0207] In certain other embodiments, the invention features a composition comprising an siNA molecule of the invention formulated with any of the cationic lipid formulations described in U.S. Patent Application Nos. 61/189,295, 61/204,878, 61/235,476, 61/249,807, and 61/298,022.
[0208] In other embodiments, the invention features conjugates and/or complexes of siNA molecules of the invention. Such conjugates and/or complexes can be used to facilitate delivery of siNA molecules into a biological system, such as a cell. The conjugates and complexes provided by the instant invention can impart therapeutic activity by transferring therapeutic compounds across cellular membranes, altering the pharmacokinetics, and/or modulating the localization of nucleic acid molecules of the invention. Non-limiting, examples of such conjugates are described in U.S. Publication Nos. US2008/0152661 Al and US 2004/0162260 Al (e.g., CDM-LBA, CDM-Pip-LBA, CDM-PEG, CDM-NAG, etc.) and U.S. Patent Application Nos. 10/427,160 and 10/201,394; and U.S. Patent Nos. 6,528,631; 6,335,434; 6,235,886; 6,153,737; 5,214,136; and 5,138,045.
[0209] In various embodiments, polyethylene glycol (PEG) can be covalently attached to siNA compounds of the present invention. The attached PEG can be any molecular weight, preferably from about 100 to about 50,000 daltons (Da).
[0210] In yet other embodiments, the invention features compositions or formulations comprising surface-modified liposomes containing poly (ethylene glycol) lipids (PEG-modified, or long-circulating liposomes or stealth liposomes) and siNA molecules of the invention, such as is disclosed in for example, International PCT Publication No. WO 96/10391; Ansell et al., International PCT Publication No. WO 96/10390; Holland et al, International PCT Publication No. WO 96/10392.
[0211] In some embodiments, the siNA molecules of the invention can also be formulated or complexed with polyethyleneimine and derivatives thereof, such as polyethyleneimine- polyethyleneglycol-N-acetylgalactosamine (PEI-PEG-GAL) or polyethyleneimine- polyethyleneglycol-tri-N-acetylgalactosamine (PEI-PEG-triGAL) derivatives. In one embodiment, the nucleic acid molecules of the invention are formulated as described in U.S. Patent Application Publication No. 20030077829. SIR-ONC-00001
[0212] In other embodiments, siNA molecules of the invention are complexed with membrane disruptive agents such as those described in U.S. Patent Application Publication No. 20010007666. In still other embodiments, the membrane disruptive agent or agents and the siNA molecule are also complexed with a cationic lipid or helper lipid molecule, such as those lipids described in U.S. Patent No. 6,235,310.
[0213] In certain embodiments, siNA molecules of the invention are complexed with delivery systems as described in U.S. Patent Application Publication Nos. 2003077829; 20050287551 ; 20050164220; 20050191627; 20050118594; 20050153919; 20050085486; and 20030158133; and International PCT Publication Nos. WO 00/03683 and WO 02/087541.
[0214] In some embodiments, a liposomal formulation of the invention comprises an siNA molecule of the invention (e.g., siNA) formulated or complexed with compounds and compositions described in U.S. Patent Nos. 6,858,224; 6,534,484; 6,287,591 ; 6,835,395; 6,586,410; 6,858,225; 6,815,432; 6,586,001 ; 6,120,798; 6,977,223; 6,998, 115; 5,981,501 ; 5,976,567; 5,705,385; and U.S. Patent Application Publication Nos. 2006/0019912; 2006/0019258; 2006/0008909; 2005/0255153; 2005/0079212; 2005/0008689; 2003/0077829, 2005/0064595, 2005/0175682, 2005/0118253; 2004/0071654; 2005/0244504; 2005/0265961 and 2003/0077829.
[0215] Alternatively, recombinant plasmids and viral vectors, as discussed above, which express siNAs of the invention can be used to deliver the molecules of the invention. Delivery of siNA molecule expressing vectors can be systemic, such as by intravenous or intra-muscular administration, by administration to target cells ex-planted from a subject followed by reintroduction into the subject, or by any other means that would allow for introduction into the desired target cell (for a review see Couture et ah, 1996, TIG., 12, 510). Such recombinant plasmids can also be administered directly or in conjunction with a suitable delivery reagents, including, for example, the Minis Transit LT1 lipophilic reagent; lipofectin; lipofectamine; cellfectin; polycations (e.g., polylysine) or liposomes lipid-based carrier system, cationic lipid, or liposome nucleic acid complexes, a micelle, a virosome, a lipid nanoparticle.
E. Kits SIR-ONC-00001
[0216] The present invention also provides nucleic acids in kit form. The kit may comprise a container. The kit typically contains a nucleic acid of the invention with instructions for its administration. In certain instances, the nucleic acids may have a targeting moiety attached. Methods of attaching targeting moieties (e.g. antibodies, proteins) are known to those of skill in the art. In certain instances, the nucleic acids are chemically modified. In other embodiments, the kit contains more than one siNA molecule of the invention. The kits may comprise an siNA molecule of the invention with a pharmaceutically acceptable carrier or diluent. The kits may further comprise excipients.
F. Therapeutic Uses/Pharmaceutical Compositions
[0217] The present body of knowledge in MET research indicates the need for methods to assay MET activity and for compounds that can regulate MET expression for research, diagnostic, and therapeutic use. As described infra, the nucleic acid molecules of the present invention can be used in assays to diagnose disease state related of MET levels. In addition, the nucleic acid molecules and pharmaceutical compositions can be used to treat disease states related to MET RNA levels.
1. Disease States Associated with MET
[0218] Particular disease states that can be associated with MET expression modulation include various cancers including solid tumors such as skin, breast, brain, cervical carcinomas, testicular carcinomas, etc. Non-limiting examples of such cancers include: Cardiac: sarcoma fibrosarcoma, rhabdomyosarcoma, and liposarcoma; Lun : bronchogenic carcinoma, squamous cell carcinoma, undifferentiated small cell carcinoma, undifferentiated large cell carcinoma, lung adenocarcinoma, alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous harmartoma, mesothelioma, non-small cell lung carcinoma, small cell lung cancer, and lung carcinoma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma), stomach (carcinoma, leiomyosarcoma), pancreatic cancer (ductal adenocarcinoma, gastrinoma), rectal cancer, colorectal carcinoma, colon cancer, gastric cancer, squamous cell carcinoma, gastric carcinoma, gastric adenocarcinomas, gastrinoma, and gall bladder carcinoma; Genitourinary tract: kidney carcinoma, bladder carcinoma, urethra, prostate (carcinoma, sarcoma, leiomyosarcoma), testis (seminoma, teratoma, embryonal SIR-ONC-00001 carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma) clear cell carcinoma, papillary renal cell carcinoma, Wilms' Tumor, and Cholangiocarcinoma; Liver: hepatoma, hepatocellular carcinoma, cholangiocarcinoma, hepatoblastoma, and liver carcinoma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, multiple myeloma, malignant giant cell tumor benign chondroma, giant cell tumors, osteosarcoma (OS), chordoma, Synovial Sarcoma, and Clear-cell sarcoma; Nervous system: meninges (meningioma, gliomatosis), brain (astrocytoma, glioblastoma), spinal cord (neurofibroma, meningioma, glioma), neuroblastoma, meningioma, neurofibroma, schwannoma, and esophogeal carcinoma; Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, cervical adenocarcinoma, pre-tumor cervical dysplasia), and ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma]); Hematologic: Adult T Cell Leukemia, Lymphomas, Multiple Myeloma, Diffuse large B-cell lymphoma, histiocytic lymphoma, Acute Myelogenous Leukemia, and Chronic Myeloid Leukemia; Skin: melanoma (malignant), basal cell carcinoma, Kaposi's sarcoma, head and neck squamous cell carcinomas, head and neck carcinoma; Breast: breast carcinoma and ductal adenocarcinoma; Thyroid: Nasopharyngeal carcinoma and thyroid carcinoma; Musculoskeletal sarcomas: Osteosarcoma, Rhabdomyosarcoma, and Synovial Sarcoma; Soft tissue sarcomas: Kaposi's Sarcoma, Leimyosarcoma, and MFH/Fibrosarcoma; Metastasis: ovarian cancer, childhood hepatocellular carcinoma, metastatic head and neck squamous cell carcinomas, gastric cancers, breast cancer, colorectal cancer, cervical cancer, lung cancer, nasopharyngeal cancer, and glioblastoma and sarcomas.
[0219] It is understood that the siNA molecules of the invention can degrade the target MET mRNA (and thus inhibit the diseases stated above). Inhibition of a disease can be evaluated by directly measuring the progress of the disease in a subject. It can also be inferred through observing a change or reversal in a condition associated with the disease. Additionally, the siNA molecules of the invention can be used as a prophylaxis. Thus, the use of the nucleic acid molecules and pharmaceutical compositions of the invention can be used to ameliorate, treat, prevent, and/or cure these diseases and others associated with regulation of MET gene expression. SIR-ONC-00001
2. Pharmaceutical Compositions
[0220] The siNA molecules of the instant invention provide useful reagents and methods for a variety of therapeutic, prophylactic, cosmetic, veterinary, diagnostic, target validation, genomic discovery, genetic engineering, and pharmacogenomic applications. a. Formulations
[0221] Thus, the present invention, in one aspect, also provides for pharmaceutical compositions of the siNA molecules described, i.e., compositions in a pharmaceutically acceptable carrier or diluent. These pharmaceutical compositions include salts of the above compounds, e.g., acid addition salts, for example, salts of hydrochloric, hydrobromic, hydroiodic, acetic acid, and benzene sulfonic acid. Other salts include for example, sodium, potassium, manganese, ammonium, and calcium salts. These formulations or compositions can comprise a pharmaceutically acceptable carrier or diluent as is generally known in the art.
[0222] In one embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1. In another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1049. In yet another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 43. In still another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1091. In another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 51. In another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1099. In another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 53. In yet another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1101. In still another embodiment, the invention features a pharmaceutical composition comprising an siNA molecule comprising formula (A). SIR-ONC-00001
[0223] The siNA molecules of the invention are preferably formulated as pharmaceutical compositions prior to administering to a subject, according to techniques known in the art. Pharmaceutical compositions of the present invention are characterized as being at least sterile and pyrogen-free. Methods for preparing pharmaceutical composition of the invention are within the skill in the art for example as described in Remington's Pharmaceutical Science, 17th ed., Mack Publishing Company, Easton, Pa. (1985).
[0224] In some embodiments, pharmaceutical compositions of the invention (e.g. siNA and/or LNP formulations thereof) further comprise conventional pharmaceutical excipients and/or additives. Suitable pharmaceutical excipients include preservatives, flavoring agents, stabilizers, antioxidants, osmolality adjusting agents, buffers, and pH adjusting agents. Suitable additives include physiologically biocompatible buffers (e.g., trimethylamine hydrochloride), addition of chelants (such as, for example, DTPA or DTPA-bisamide) or calcium chelate complexes (as for example calcium DTPA, CaNaDTPA-bisamide), or, optionally, additions of calcium or sodium salts (for example, calcium chloride, calcium ascorbate, calcium gluconate or calcium lactate). In addition, antioxidants and suspending agents can be used.
[0225] Non-limiting examples of various types of formulations for local administration include ointments, lotions, creams, gels, foams, preparations for delivery by transdermal patches, powders, sprays, aerosols, capsules or cartridges for use in an inhaler or insufflator or drops (for example eye or nose drops), solutions/suspensions for nebulization, suppositories, pessaries, retention enemas and chewable or suckable tablets or pellets (for example for the treatment of aphthous ulcers) or liposome or microencapsulation preparations.
[0226] Ointments, creams and gels, can, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agent and/or solvents. Non limiting examples of such bases can thus, for example, include water and/or an oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil, or a solvent such as polyethylene glycol. Thickening agents and gelling agents which can be used according to the nature of the base. Non-limiting examples of such agents include soft paraffin, aluminum stearate, cetostearyl alcohol, polyethylene glycols, woolfat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents. SIR-ONC-00001
[0227] In one embodiment lotions can be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents or thickening agents.
[0228] In one embodiment powders for external application can be formed with the aid of any suitable powder base, for example, talc, lactose or starch. Drops can be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilizing agents, suspending agents or preservatives.
[0229] Compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients can be, for example, inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets can be uncoated or they can be coated by known techniques. In some cases such coatings can be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monosterate or glyceryl distearate can be employed.
[0230] Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
[0231] Aqueous suspensions contain the active materials in a mixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents can SIR-ONC-00001 be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate; or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions can also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
[0232] Oily suspensions can be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions can contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents and flavoring agents can be added to provide palatable oral preparations. These compositions can be preserved by the addition of an antioxidant such as ascorbic acid
[0233] Pharmaceutical compositions of the invention can also be in the form of oil-in-water emulsions. The oily phase can be a vegetable oil or a mineral oil or mixtures of these. Suitable emulsifying agents can be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions can also contain sweetening and flavoring agents.
[0234] Syrups and elixirs can be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations can also contain a demulcent, a preservative and flavoring and coloring agents. The pharmaceutical compositions can be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above. The sterile injectable preparation can also SIR-ONC-00001 be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil can be employed including synthetic mono-or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
[0235] The nucleic acid molecules of the invention can also be administered in the form of suppositories, e.g., for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter and polyethylene glycols.
[0236] Nucleic acid molecules of the invention can be administered parenterally in a sterile medium. The drug, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle.
[0237] In other embodiments, the siNA and LNP compositions and formulations provided herein for use in pulmonary delivery further comprise one or more surfactants. Suitable surfactants or surfactant components for enhancing the uptake of the compositions of the invention include synthetic and natural as well as full and truncated forms of surfactant protein A, surfactant protein B, surfactant protein C, surfactant protein D and surfactant Protein E, di- saturated phosphatidylcholine (other than dipalmitoyl), dipalmitoylphosphatidylcholine, phosphatidylcholine, phosphatidylglycerol, phosphatidylinositol, phosphatidylethanolamine, phosphatidylserine; phosphatidic acid, ubiquinones, lysophosphatidylethanolamine, lysophosphatidylcholine, palmitoyl-lysophosphatidylcholine, dehydroepiandrosterone, dolichols, sulfatidic acid, glycerol-3-phosphate, dihydroxyacetone phosphate, glycerol, glycero-3- phosphocholine, dihydroxyacetone, palmitate, cytidine diphosphate (CDP) diacylglycerol, CDP choline, choline, choline phosphate; as well as natural and artificial lamellar bodies which are the natural carrier vehicles for the components of surfactant, omega-3 fatty acids, polyenic acid, polyenoic acid, lecithin, palmitinic acid, non-ionic block copolymers of ethylene or propylene SIR-ONC-00001 oxides, polyoxypropylene, monomeric and polymeric, polyoxyethylene, monomeric and polymeric, poly (vinyl amine) with dextran and/or alkanoyl side chains, Brij 35, Triton X-100 and synthetic surfactants ALEC, Exosurf, Survan and Atovaquone, among others. These surfactants can be used either as single or part of a multiple component surfactant in a formulation, or as covalently bound additions to the 5' and/or 3' ends of the nucleic acid component of a pharmaceutical composition herein. b. Combinations
[0238] The siNAs and pharmaceutical formulations according to the invention can be administered to a subject alone or used in combination with or include one or more other therapeutic agents, for example, anticancer agents. Thus, combinations of the presently disclosed compounds with other anti-cancer or chemotherapeutic agents are within the scope of the invention. Examples of such agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S. Hellman (editors), 6th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved. Such anti-cancer agents include, but are not limited to, the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic/cytostatic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG- CoA reductase inhibitors and other angiogenesis inhibitors, inhibitors of cell proliferation and survival signaling, apoptosis inducing agents and agents that interfere with cell cycle checkpoints. The siNAs of the invention are also useful in combination with any therapeutic agent used in the treatment of HCC, for example, but not limitation sorafenib. The instant compounds are particularly useful when co-administered with radiation therapy.
[0239] In a further embodiment, therefore, the invention provides a combination comprising an siNA molecule of the invention, such as for example, but not limitation, an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A) or a pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof together with one or more anti-cancer or chemotherapeutic agents. SIR-ONC-00001
[0240] In certain embodiments, the instant siNA molecules of the invention are useful in combination with known anti-cancer agents including the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors, HIV protease inhibitors, reverse transcriptase inhibitors, and other angiogenesis inhibitors.
[0241] Examples of estrogen receptor modulators that can be used in combination with the compounds of the invention include, but are not limited to, tamoxifen, raloxifene, idoxifene, LY353381, LY117081, toremifene, fulvestrant, 4-[7-(2,2-dimethyl-l-oxopropoxy-4-methyl-2- [4- [2- ( 1 -piperidinyl)ethoxy] phenyl] -2H- 1 -benzopyran-3-yl] -phenyl-2,2-dimethylpropanoate, 4,4' -dihydroxybenzophenone-2,4-dinitrophenyl-hydrazone, and SH646.
[0242] Examples of androgen receptor modulators that can be used in combination with the compounds of the invention include, but are not limited to, finasteride and other 5a-reductase inhibitors, nilutamide, flutamide, bicalutamide, liarozole, and abiraterone acetate.
[0243] Examples of such retinoid receptor modulators that can be used in combination with the compounds of the invention include, but are not limited to, bexarotene, tretinoin, 13-cis- retinoic acid, 9-cis-retinoic acid, -difluoromethylornithine, ILX23-7553, trans-N-(4'- hydroxyphenyl) retinamide, and N-4-carboxyphenyl retinamide.
[0244] Examples of cytotoxic agents that can be used in combination with the compounds of the invention include, but are not limited to, sertenef, cachectin, ifosfamide, tasonermin, lonidamine, carboplatin, altretamine, prednimustine, dibromodulcitol, ranimustine, fotemustine, nedaplatin, oxaliplatin, temozolomide, heptaplatin, estramustine, improsulfan tosilate, trofosfamide, nimustine, dibrospidium chloride, pumitepa, lobaplatin, satraplatin, profiromycin, cisplatin, irofulven, dexifosfamide, cis-aminedichloro(2-methyl-pyridine)platinum, benzylguanine, glufosfamide, GPX100, (trans, trans, trans)-bis-mu-(hexane-l,6-diamine)-mu- [diamine-platinum(II)]bis[diamine(chloro)platinum (II)] tetrachloride, diarizidinylspermine, arsenic trioxide, l-(l l-dodecylamino-10-hydroxyundecyl)-3,7-dimethylxanthine, zorubicin, idarubicin, daunorubicin, bisantrene, mitoxantrone, pirarubicin, pinafide, valrubicin, amrubicin, antineoplaston, 3 ' -deamino-3 ' -morpholino- 13-deoxo- 10-hydroxycarminomycin, annamycin, SIR-ONC-00001 galarubicin, elinafide, MEN10755, and 4-demethoxy-3-deamino-3-aziridinyl-4- methylsulphonyl-daunorubicin (see WO 00/50032).
[0245] An example of a hypoxia activatable compound that can be used in combination with the compounds of the invention is tirapazamine.
[0246] Examples of proteasome inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, lactacystin and bortezomib.
[0247] Examples of microtubule inhibitors/microtubule-stabilising agents that can be used in combination with the compounds of the invention include, but are not limited to, paclitaxel, vindesine sulfate, 3',4'-didehydro-4'-deoxy-8'-norvincaleukoblastine, docetaxol, rhizoxin, dolastatin, mivobulin isethionate, auristatin, cemadotin, RPR109881, BMS 184476, vinflunine, cryptophycin, 2,3,4,5, 6-pentafluoro-N-(3-fluoro-4-methoxyphenyl) benzene sulfonamide, anhydrovinblastine, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-prolyl-L-proline-t- butylamide, TDX258, the epothilones (see for example U.S. Pat. Nos. 6,284,781 and 6,288,237) and BMS 188797.
[0248] Some examples of topoisomerase inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, are topotecan, hycaptamine, irinotecan, rubitecan, 6-ethoxypropionyl-3',4'-0-exo-benzylidene-chartreusin, 9-methoxy-N,N- dimethyl-5-nitropyrazolo[3,4,5-kl]acridine-2-(6H) propanamine, l-amino-9-ethyl-5-fluoro-2,3- dihydro-9 -hydroxy-4-methyl- 1 H, 12H-benzo [de]pyrano [3 ' ,4' :b,7] -indolizino [ 1 ,2b] quinoline- 10,13(9H,15H)dione, lurtotecan, 7-[2-(N-isopropylamino)ethyl]-(20S)camptothecin, BNP1350, BNPI1100, BN80915, BN80942, etoposide phosphate, teniposide, sobuzoxane, 2'- dimethylamino-2'-deoxy-etoposide, GL331, N-[2-(dimethylamino)ethyl]-9-hydroxy-5,6- dimethyl-6H-pyrido[4,3-b]carbazole-l-carboxamide, asulacrine, (5a, 5aB, 8aa,9b)-9-[2-[N-[2- (dimethylamino)ethyl]-N-methylamino]ethyl]-5-[4-hydro0xy-3,5-dimethoxyphenyl]- 5,5a,6,8,8a,9-hexohydrofuro(3',4':6,7)naphtho(2,3-d)-l,3-dioxol-6-one, 2,3-(methylenedioxy)-5- methyl-7-hydroxy-8-methoxybenzo[c]-phenanthridinium, 6,9-bis[(2- aminoethyl)amino]benzo[g]isoguinoline-5, 10-dione, 5-(3-aminopropylamino)-7, 10-dihydroxy-2- (2-hydroxyethylaminomethyl)-6H-pyrazolo[4,5,l-de]acridin-6-one, N-[l- [2(diethylamino)ethylamino]-7-methoxy-9-oxo-9H-thioxanthen-4-ylmethyl]formamide, N-(2- SIR-ONC-00001
(dimethylamino)ethyl)acridine-4-carboxamide, 6-[[2-(dimethylamino)ethyl]amino]-3-hydroxy- 7H-indeno[2,l-c] quinolin-7-one, and dimesna.
[0249] Examples of inhibitors of mitotic kinesins, and in particular the human mitotic kinesin KSP, that can be used in combination with the compounds of the invention include, but are not limited to, inhibitors described in PCT Publications WO 01/30768, WO 01/98278, WO 03/050,064, WO 03/050,122, WO 03/049,527, WO 03/049,679, WO 03/049,678, WO04/039774, WO03/079973, WO03/099211, WO03/105855, WO03/106417, WO04/037171, WO04/058148, WO04/058700, WO04/126699, WO05/018638, WO05/019206, WO05/019205, WO05/018547, WO05/017190, US2005/0176776. In an embodiment inhibitors of mitotic kinesins include, but are not limited to inhibitors of KSP, inhibitors of MKLPl, inhibitors of CENP-E, inhibitors of MCAK, inhibitors of Kifl4, inhibitors of Mphosphl and inhibitors of Rab6-KIFL.
[0250] Examples of "histone deacetylase inhibitors" that can be used in combination with the compounds of the invention include, but are not limited to, TSA, oxamflatin, PXD101, MG98, valproic acid and scriptaid. Further reference to other histone deacetylase inhibitors may be found in the following manuscript; Miller, T.A. et al. J. Med. Chem. 46(24):5097-5116 (2003).
[0251] Inhibitors of kinases involved in mitotic progression that can be used in combination with the compounds of the invention include, but are not limited to, inhibitors of aurora kinase, inhibitors of Polo-like kinases (PLK) (in particular inhibitors of PLK-1), inhibitors of bub-1 and inhibitors of bub-Rl.
[0252] Antiproliferative agents that can be used in combination with the compounds of the invention include, but are not limited to, antisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231, and INX3001, and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fludarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2'-deoxy-2'-methylidenecytidine, 2'- fluoromethylene-2'-deoxycytidine, N-[5-(2,3-dihydro-benzofuryl)sulfonyl]-N'-(3,4- dichlorophenyl)urea, N6-[4-deoxy-4-[N2-[2(E),4(E)-tetradecadienoyl]glycylamino]-L-glycero- B-L-manno-heptopyranosyl]adenine, aplidine, ecteinascidin, troxacitabine, 4-[2-amino-4-oxo- 4,6,7, 8-tetrahydro-3H-pyrimidino[5,4-b][l,4]thiazin-6-yl-(S)-ethyl]-2,5-thienoyl-L-glutamic SIR-ONC-00001 acid, aminopterin, 5-flurouracil, alanosine, 1 l-acetyl-8-(carbamoyloxymethyl)-4-formyl-6- methoxy-14-oxa-l,l l-diazatetracyclo(7.4.1.0.0)-tetradeca-2,4,6-trien-9-yl acetic acid ester, swainsonine, lometrexol, dexrazoxane, methioninase, 2'-cyano-2'-deoxy-N4-palmitoyl-l-B-D- arabino furanosyl cytosine and 3-aminopyridine-2-carboxaldehyde thiosemicarbazone.
[0253] Examples of monoclonal antibody targeted therapeutic agents that can be used in combination with the compounds of the invention include those therapeutic agents which have cytotoxic agents or radioisotopes attached to a cancer cell specific or target cell specific monoclonal antibody, such as, for example, Bexxar.
[0254] Examples of HMG-CoA reductase inhibitors that may be used that can be used in combination with the compounds of the invention include, but are not limited to, lovastatin
(MEVACOR®; see U.S. Pat. Nos. 4,231,938, 4,294,926 and 4,319,039), simvastatin (ZOCOR®; see U.S. Pat. Nos. 4,444,784, 4,820,850 and 4,916,239), pravastatin (PRAVACHOL®; see U.S.
Pat. Nos. 4,346,227, 4,537,859, 4,410,629, 5,030,447 and 5,180,589), fluvastatin (LESCOL®; see U.S. Pat. Nos. 5,354,772, 4,911,165, 4,929,437, 5,189,164, 5,118,853, 5,290,946 and
5,356,896) and atorvastatin (LIPITOR®; see U.S. Pat. Nos. 5,273,995, 4,681,893, 5,489,691 and 5,342,952). The structural formulas of these and additional HMG-CoA reductase inhibitors that may be used in the instant methods are described at page 87 of M. Yalpani, "Cholesterol Lowering Drugs", Chemistry & Industry, pp. 85-89 (5 February 1996) and US Patent Nos. 4,782,084 and 4,885,314.
[0255] Examples of prenyl-protein transferase inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, can be found in the following publications and patents: WO 96/30343, WO 97/18813, WO 97/21701, WO 97/23478, WO 97/38665, WO 98/28980, WO 98/29119, WO 95/32987, U.S. Pat. No. 5,420,245, U.S. Pat. No. 5,523,430, U.S. Pat. No. 5,532,359, U.S. Pat. No. 5,510,510, U.S. Pat. No. 5,589,485, U.S. Pat. No. 5,602,098, European Patent Publ. 0 618 221, European Patent Publ. 0 675 112, European Patent Publ. 0 604 181, European Patent Publ. 0 696 593, WO 94/19357, WO 95/08542, WO 95/11917, WO 95/12612, WO 95/12572, WO 95/10514, U.S. Pat. No. 5,661,152, WO 95/10515, WO 95/10516, WO 95/24612, WO 95/34535, WO 95/25086, WO 96/05529, WO 96/06138, WO 96/06193, WO 96/16443, WO 96/21701, WO 96/21456, WO 96/22278, WO SIR-ONC-00001
96/24611, WO 96/24612, WO 96/05168, WO 96/05169, WO 96/00736, U.S. Pat. No. 5,571,792, WO 96/17861, WO 96/33159, WO 96/34850, WO 96/34851, WO 96/30017, WO 96/30018, WO 96/30362, WO 96/30363, WO 96/31111, WO 96/31477, WO 96/31478, WO 96/31501, WO 97/00252, WO 97/03047, WO 97/03050, WO 97/04785, WO 97/02920, WO 97/17070, WO 97/23478, WO 97/26246, WO 97/30053, WO 97/44350, WO 98/02436, and U.S. Pat. No. 5,532,359. For an example of the role of a prenyl-protein transferase inhibitor on angiogenesis see European J. of Cancer, Vol. 35, No. 9, pp.1394-1401 (1999).
[0256] Examples of angiogenesis inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, tyrosine kinase inhibitors, such as inhibitors of the tyrosine kinase receptors Flt-1 (VEGFR1) and Flk-l/KDR (VEGFR2), inhibitors of epidermal-derived, fibroblast-derived, or platelet derived growth factors, MMP (matrix metalloprotease) inhibitors, integrin blockers, interferon- , interleukin-12, pentosan polysulfate, cyclooxygenase inhibitors, including nonsteroidal anti-inflammatories (NSAIDs) like aspirin and ibuprofen as well as selective cyclooxy-genase-2 inhibitors like celecoxib and rofecoxib (PNAS, Vol. 89, p. 7384 (1992); JNCI, Vol. 69, p. 475 (1982); Arch. OpthalmoL, Vol. 108, p.573 (1990); Anat. Rec, Vol. 238, p. 68 (1994); FEBS Letters, Vol. 372, p. 83 (1995); Clin, Orthop. Vol. 313, p. 76 (1995); J. Mol. Endocrinol., Vol. 16, p.107 (1996); Jpn. J. Pharmacol., Vol. 75, p. 105 (1997); Cancer Res., Vol. 57, p. 1625 (1997); Cell, Vol. 93, p. 705 (1998); Intl. J. Mol. Med., Vol. 2, p. 715 (1998); J. Biol. Chem., Vol. 274, p. 9116 (1999)), steroidal anti-inflammatories (such as corticosteroids, mineralocorticoids, dexamethasone, prednisone, prednisolone, methylpred, betamethasone), carboxyamidotriazole, combretastatin A-4, squalamine, 6-0- chloroacetyl-carbonyl)-fumagillol, thalidomide, angiostatin, troponin- 1, angiotensin II antagonists (see Fernandez et al., J. Lab. Clin. Med. 105: 141-145 (1985)), and antibodies to VEGF (see, Nature Biotechnology, Vol. 17, pp.963-968 (October 1999); Kim et al., Nature, 362, 841-844 (1993); WO 00/44777; and WO 00/61186).
[0257] Other therapeutic agents that modulate or inhibit angiogenesis may also be used in combination with the compounds of the instant inventionand include agents that modulate or inhibit the coagulation and fibrinolysis systems (see review in Clin. Chem. La. Med. 38:679-692 (2000)). Examples of such agents that modulate or inhibit the coagulation and fibrinolysis pathways that can be used in combination with the compounds of the invention include, but are SIR-ONC-00001 not limited to, heparin (see Thromb. Haemost. 80: 10-23 (1998)), low molecular weight heparins and carboxypeptidase U inhibitors (also known as inhibitors of active thrombin activatable fibrinolysis inhibitor [TAFIa]) (see Thrombosis Res. 101:329-354 (2001)). TAFIa inhibitors have been described in PCT Publication WO 03/013,526 and U,S, Ser. No. 60/349,925 (filed January 18, 2002).
[0258] Agents that interfere with cell cycle checkpoints that can be used in combination with the compounds of the invention include, but are not limited to, inhibitors of ATR, ATM, the Chkl and Chk2 kinases and cdk and cdc kinase inhibitors and are specifically exemplified by 7- hydroxystaurosporin, flavopiridol, CYC202 (Cyclacel) and BMS-387032.
[0259] Agents that interfere with receptor tyrosine kinases (RTKs) that can be used in combination with the compounds of the invention include, but are not limited to, inhibitors of c- Kit, Eph, PDGF, Flt3 and MET. Further agents include inhibitors of RTKs as described by Bume- Jensen and Hunter, Nature, 411:355-365, 2001.
[0260] Inhibitors of cell proliferation and survival signaling pathway that can be used in combination with the compounds of the invention include, but are not limited to, inhibitors of EGFR (for example gefitinib and erlotinib), inhibitors of ERB-2 (for example trastuzumab), inhibitors of IGFR, inhibitors of cytokine receptors, inhibitors of MET, inhibitors of PI3K (for example LY294002), serine/threonine kinases (including but not limited to inhibitors of Akt such as described in WO 02/083064, WO 02/083139, WO 02/083140, US 2004-0116432, WO 02/083138, US 2004-0102360, WO 03/086404, WO 03/086279, WO 03/086394, WO 03/084473, WO 03/086403, WO 2004/041162, WO 2004/096131, WO 2004/096129, WO 2004/096135, WO 2004/096130, WO 2005/100356, WO 2005/100344), inhibitors of Raf kinase (for example BAY-43-9006), inhibitors of MEK (for example CI- 1040 and PD-098059) and inhibitors of mTOR (for example Wyeth CCT779). Such agents include small molecule inhibitor compounds and antibody antagonists.
[0261] Apoptosis inducing agents that can be used in combination with the compounds of the invention include, but are not limited to, activators of TNF receptor family members (including the TRAIL receptors). SIR-ONC-00001
[0262] NSAIDs that are selective COX-2 inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, those NSAIDs disclosed in U.S. Pat. 5,474,995, U.S. Pat. 5,861,419, U.S. Pat. 6,001,843, U.S. Pat. 6,020,343, U.S. Pat. 5,409,944, U.S. Pat. 5,436,265, U.S. Pat. 5,536,752, U.S. Pat. 5,550, 142, U.S. Pat. 5,604,260, U.S. 5,698,584, U.S. Pat. 5,710,140, WO 94/15932, U.S. Pat. 5,344,991, U.S. Pat. 5, 134,142, U.S. Pat. 5,380,738, U.S. Pat. 5,393,790, U.S. Pat. 5,466,823, U.S. Pat. 5,633,272, and U.S. Pat. 5,932,598, all of which are hereby incorporated by reference.
[0263] Inhibitors of COX-2 that are particularly useful in combination with the compounds of the invention include: 3-phenyl-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; and 5-chloro- 3-(4-methylsulfonyl)-phenyl-2-(2-methyl-5-pyridinyl)pyridine; or a pharmaceutically acceptable salt thereof.
[0264] Compounds that have been described as specific inhibitors of COX-2 and are therefore useful in the present invention include, but are not limited to: parecoxib, CELEBREX® and BEXTRA® or a pharmaceutically acceptable salt thereof.
[0265] Angiogenesis inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, endostatin, ukrain, ranpirnase, IM862, 5-methoxy-4-[2- methyl-3-(3-methyl-2-butenyl)oxiranyl]- l-oxaspiro[2,5]oct-6-yl(chloroacetyl)carbamate, acetyldinanaline, 5-amino- l-[[3,5-dichloro-4-(4-chlorobenzoyl)-phenyl]methyl]-lH- l,2,3- triazole-4-carboxamide,CM101, squalamine, combretastatin, RPI4610, NX31838, sulfated mannopentaose phosphate, 7,7-(carbonyl-bis[imino-N-methyl-4,2-pyrrolocarbonylimino[N- methyl-4,2-pyrrole]-carbonylimino]-bis-(l,3-naphthalene disulfonate), and 3-[(2,4- dimethylpyrrol-5-yl)methylene]-2-indolinone (SU5416).
[0266] Tyrosine kinase inhibitors that can be used in combination with the compounds of the invention include, but are not limited to, N-(trifluoromethylphenyl)-5-methylisoxazol-4- carboxamide, 3-[(2,4-dimethylpyrrol-5-yl)methylidenyl)indolin-2-one, 17-(allylamino)- 17- demethoxygeldanamycin, 4-(3-chloro-4-fluorophenylamino)-7-methoxy-6-[3-(4- morpholinyl)propoxyl]quinazoline, N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4- quinazolinamine, BIBX1382, 2,3,9, 10,11, 12-hexahydro- 10-(hydroxymethyl)- 10-hydroxy-9- methyl-9,12-epoxy- lH-diindolo[l,2,3-fg:3',2', -kl]pyrrolo[3,4-i] [l,6]benzodiazocin- l-one, SIR-ONC-00001
SH268, genistein, imatinib (STI571), CEP2563, 4-(3-chlorophenylamino)-5,6-dimethyl-7H- pyrrolo[2,3-d]pyrimidinemethane sulfonate, 4-(3-bromo-4-hydroxyphenyl)amino-6,7- dimethoxyquinazoline, 4-(4' -hydroxyphenyl)amino-6,7-dimethoxyquinazoline, SU6668, STI571A, N-4-chlorophenyl-4-(4-pyridylmethyl)-l-phthalazinamine, and EMD121974.
[0267] Combinations with compounds other than anti-cancer compounds are also encompassed in the instant compositions and methods. For example, combinations of the instantly claimed compounds with PPAR-γ (i.e., PPAR-gamma) agonists and PPAR-δ (i.e., PPAR-delta) agonists are useful in the treatment of certain malingnancies. PPAR-y and PPAR-δ are the nuclear peroxisome proliferator-activated receptors γ and δ. The expression of PPAR-γ on endothelial cells and its involvement in angiogenesis has been reported in the literature (see J. Cardiovasc. Pharmacol. 1998; 31:909-913; J. Biol. Chem. 1999;274:9116-9121; Invest. Ophthalmol Vis. Sci. 2000; 41:2309-2317). More recently, PPAR-γ agonists have been shown to inhibit the angiogenic response to VEGF in vitro; both troglitazone and rosiglitazone maleate inhibit the development of retinal neovascularization in mice. (Arch. Ophthamol. 2001; 119:709-717). Examples of PPAR-γ agonists and PPAR- γ/ agonists that can be used in combination with the compounds of the invention include, but are not limited to, thiazolidinediones (such as DRF2725, CS-011, troglitazone, rosiglitazone, and pioglitazone), fenofibrate, gemfibrozil, clofibrate, GW2570, SB219994, AR-H039242, JTT-501, MCC-555, GW2331, GW409544, NN2344, KRP297, NP0110, DRF4158, NN622, GI262570, PNU182716, DRF552926, 2-[(5,7-dipropyl-3-trifluoromethyl-l,2-benzisoxazol-6-yl)oxy]-2-methylpropionic acid (disclosed in USSN 09/782,856), and 2(R)-7-(3-(2-chloro-4-(4-fluorophenoxy) phenoxy)propoxy)-2-ethylchromane-2-carboxylic acid (disclosed in USSN 60/235,708 and 60/244,697).
[0268] Another embodiment of the instant invention is the use of the presently disclosed compounds in combination with gene therapy for the treatment of cancer. For an overview of genetic strategies to treating cancer see Hall et al. (Am J Hum Genet 61:785-789, 1997) and Kufe et al. (Cancer Medicine, 5th Ed, pp 876-889, BC Decker, Hamilton 2000). Gene therapy can be used to deliver any tumor suppressing gene. Examples of such genes include, but are not limited to, p53, which can be delivered via recombinant virus -mediated gene transfer (see U.S. Pat. No. 6,069,134, for example), a uPA/uPAR antagonist ("Adenovirus-Mediated Delivery of a SIR-ONC-00001 uPA/uPAR Antagonist Suppresses Angiogenesis-Dependent Tumor Growth and Dissemination in Mice," Gene Therapy, August 1998;5(8): 1105-13), and interferon gamma (/ Immunol 2000;164:217-222).
[0269] The compounds of the instant invention may also be administered in combination with an inhibitor of inherent multidrug resistance (MDR), in particular MDR associated with high levels of expression of transporter proteins. Such MDR inhibitors include inhibitors of p- glycoprotein (P-gp), such as LY335979, XR9576, OC144-093, R101922, VX853 and PSC833 (valspodar).
[0270] A compound of the present invention may be employed in conjunction with antiemetic agents to treat nausea or emesis, including acute, delayed, late -phase, and anticipatory emesis, which may result from the use of a compound of the present invention, alone or with radiation therapy. For the prevention or treatment of emesis, a compound of the present invention may be used in conjunction with other anti-emetic agents, especially neurokinin- 1 receptor antagonists, 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, a corticosteroid such as Decadron (dexamethasone), Kenalog, Aristocort, Nasalide, Preferid, Benecorten or others such as disclosed in U.S.Patent Nos. 2,789,118, 2,990,401, 3,048,581, 3,126,375, 3,929,768, 3,996,359, 3,928,326 and 3,749,712, an antidopaminergic, such as the phenothiazines (for example prochlorperazine, fluphenazine, thioridazine and mesoridazine), metoclopramide or dronabinol. In an embodiment, an anti-emesis agent selected from a neurokinin- 1 receptor antagonist, a 5HT3 receptor antagonist and a corticosteroid is administered as an adjuvant for the treatment or prevention of emesis that may result upon administration of the instant compounds.
[0271] Neurokinin- 1 receptor antagonists of use in conjunction with the compounds of the present invention are fully described, for example, in U.S. Pat. Nos. 5,162,339, 5,232,929, 5,242,930, 5,373,003, 5,387,595, 5,459,270, 5,494,926, 5,496,833, 5,637,699, 5,719,147; European Patent Publication Nos. EP 0 360 390, 0 394 989, 0 428 434, 0 429 366, 0 430 771, 0 436 334, 0 443 132, 0 482 539, 0 498 069, 0 499 313, 0 512 901, 0 512 902, 0 514 273, 0 514 274, 0 514 275, 0 514 276, 0 515 681, 0 517 589, 0 520 555, 0 522 808, 0 528 495, 0 532 456, 0 533 280, 0 536 817, 0 545 478, 0 558 156, 0 577 394, 0 585 913,0 590 152, 0 599 538, 0 610 SIR-ONC-00001
793, 0 634 402, 0 686 629, 0 693 489, 0 694 535, 0 699 655, 0 699 674, 0 707 006, 0 708 101, 0 709 375, 0 709 376, 0 714 891, 0 723 959, 0 733 632 and 0 776 893; PCT International Patent Publication Nos. WO 90/05525, 90/05729, 91/09844, 91/18899, 92/01688, 92/06079, 92/12151, 92/15585, 92/17449, 92/20661, 92/20676, 92/21677, 92/22569, 93/00330, 93/00331, 93/01159, 93/01165, 93/01169, 93/01170, 93/06099, 93/09116, 93/10073, 93/14084, 93/14113, 93/18023, 93/19064, 93/21155, 93/21181, 93/23380, 93/24465, 94/00440, 94/01402, 94/02461, 94/02595, 94/03429, 94/03445, 94/04494, 94/04496, 94/05625, 94/07843, 94/08997, 94/10165, 94/10167, 94/10168, 94/10170, 94/11368, 94/13639, 94/13663, 94/14767, 94/15903, 94/19320, 94/19323, 94/20500, 94/26735, 94/26740, 94/29309, 95/02595, 95/04040, 95/04042, 95/06645, 95/07886, 95/07908, 95/08549, 95/11880, 95/14017, 95/15311, 95/16679, 95/17382, 95/18124, 95/18129, 95/19344, 95/20575, 95/21819, 95/22525, 95/23798, 95/26338, 95/28418, 95/30674, 95/30687, 95/33744, 96/05181, 96/05193, 96/05203, 96/06094, 96/07649, 96/10562, 96/16939, 96/18643, 96/20197, 96/21661, 96/29304, 96/29317, 96/29326, 96/29328, 96/31214, 96/32385, 96/37489, 97/01553, 97/01554, 97/03066, 97/08144, 97/14671, 97/17362, 97/18206, 97/19084, 97/19942 and 97/21702; and in British Patent Publication Nos. 2 266 529, 2 268 931, 2 269 170, 2 269 590, 2 271 774, 2 292 144, 2 293 168, 2 293 169, and 2 302 689. The preparation of such compounds is fully described in the aforementioned patents and publications, which are incorporated herein by reference.
[0272] In an embodiment, the neurokinin- 1 receptor antagonist for use in conjunction with the compounds of the present invention is selected from: 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)- phenyl)ethoxy)-3 -(S )- (4-fluorophenyl)-4- (3- (5 -oxo- 1 H,4H- 1 ,2,4-triazolo)methyl)morpholine, or a pharmaceutically acceptable salt thereof, which is described in U.S. Pat. No. 5,719,147.
[0273] A compound of the instant invention may also be useful for treating or preventing cancer, including bone cancer, in combination with bisphosphonates (understood to include bisphosphonates, diphosphonates, bisphosphonic acids and diphosphonic acids). Examples of bisphosphonates include but are not limited to: etidronate (Didronel), pamidronate (Aredia), alendronate (Fosamax), risedronate (Actonel), zoledronate (Zometa), ibandronate (Boniva), incadronate or cimadronate, clodronate, EB-1053, minodronate, neridronate, piridronate and tiludronate including any and all pharmaceutically acceptable salts, derivatives, hydrates and mixtures thereof. SIR-ONC-00001
[0274] A compound of the instant invention may also be administered with an agent useful in the treatment of anemia. Such an anemia treatment agent is, for example, a continuous eythropoiesis receptor activator (such as epoetin alfa).
[0275] A compound of the instant invention may also be administered with an agent useful in the treatment of neutropenia. Such a neutropenia treatment agent is, for example, a hematopoietic growth factor which regulates the production and function of neutrophils such as a human granulocyte colony stimulating factor, (G-CSF). Examples of a G-CSF include filgrastim and PEG-filgrastim.
[0276] A compound of the instant invention may also be administered with an immunologic - enhancing drug, such as levamisole, isoprinosine and Zadaxin.
[0277] A compound of the instant invention may also be useful for treating or preventing breast cancer in combination with aromatase inhibitors. Examples of aromatase inhibitors include but are not limited to: anastrozole, letrozole and exemestane.
[0278] A compound of the instant invention may also be useful for treating or preventing cancer in combination with other siNA therapeutics.
[0279] The compounds of the instant invention may also be administered in combination with γ-secretase inhibitors and/or inhibitors of NOTCH signaling. Such inhibitors include compounds described in WO 01/90084, WO 02/30912, WO 01/70677, WO 03/013506, WO 02/36555, WO 03/093252, WO 03/093264, WO 03/093251, WO 03/093253, WO 2004/039800, WO 2004/039370, WO 2005/030731, WO 2005/014553, USSN 10/957,251, WO 2004/089911, WO 02/081435, WO 02/081433, WO 03/018543, WO 2004/031137, WO 2004/031139, WO 2004/031138, WO 2004/101538, WO 2004/101539 and WO 02/47671 (including LY-450139).
[0280] A compound of the instant invention may also be useful for treating or preventing cancer in combination with PARP inhibitors.
[0281] A compound of the instant invention may also be useful for treating cancer in combination with the following therapeutic agents: abarelix (Plenaxis depot®); aldesleukin (Prokine®); Aldesleukin (Proleukin®); Alemtuzumabb (Campath®); alitretinoin (Panretin®); SIR-ONC-00001 allopurinol (Zyloprim®); altretamine (Hexalen®); amifostine (Ethyol®); anastrozole (Arimidex®); arsenic trioxide (Trisenox®); asparaginase (Elspar®); azacitidine (Vidaza®); bendamustine hydrochloride (Treanda®); bevacuzimab (Avastin®); bexarotene capsules (Targretin®); bexarotene gel (Targretin®); bleomycin (Blenoxane®); bortezomib (Velcade®); brefeldin A; busulfan intravenous (Busulfex®); busulfan oral (Myleran®); calusterone (Methosarb®); capecitabine (Xeloda®); carboplatin (Paraplatin®); carmustine (BCNU®, BiCNU®); carmustine (Gliadel®); carmustine with Polifeprosan 20 Implant (Gliadel Wafer®); celecoxib (Celebrex®); cetuximab (Erbitux®); chlorambucil (Leukeran®); cisplatin (Platinol®); cladribine (Leustatin®, 2-CdA®); clofarabine (Clolar®); cyclophosphamide (Cytoxan®, Neosar®); cyclophosphamide (Cytoxan Injection®); cyclophosphamide (Cytoxan Tablet®); cytarabine (Cytosar-U®); cytarabine liposomal (DepoCyt®); dacarbazine (DTIC-Dome®); dactinomycin, actinomycin D (Cosmegen®); dalteparin sodium injection (Fragmin®); Darbepoetin alfa (Aranesp®); dasatinib (Sprycel®); daunorubicin liposomal (DanuoXome®); daunorubicin, daunomycin (Daunorubicin®); daunorubicin, daunomycin (Cerubidine®); degarelix (Firmagon®); Denileukin diftitox (Ontak®); dexrazoxane (Zinecard®); dexrazoxane hydrochloride (Totect®); didemnin B; 17-DMAG; docetaxel (Taxotere®); doxorubicin (Adriamycin PFS®); doxorubicin (Adriamycin®, Rubex®); doxorubicin (Adriamycin PFS Injection®); doxorubicin liposomal (Doxil®); dromostanolone propionate (Dromostanolone ®); dromostanolone propionate (Masterone Injection®); eculizumab injection (Soliris®); Elliott's B Solution (Elliott's B Solution®); eltrombopag (Promacta®); epirubicin (Ellence®); Epoetin alfa (epogen®); erlotinib (Tarceva®); estramustine (Emcyt®); ethinyl estradiol; etoposide phosphate (Etopophos®); etoposide, VP- 16 (Vepesid®); everolimus tablets (Afinitor®); exemestane (Aromasin®); ferumoxytol (Feraheme Injection®); Filgrastim (Neupogen®); floxuridine (intraarterial) (FUDR®); fludarabine (Fludara®); fluorouracil, 5-FU (Adrucil®); fulvestrant (Faslodex®); gefitinib (Iressa®); geldanamycin; gemcitabine (Gemzar®); gemtuzumab ozogamicin (Mylotarg®); goserelin acetate (Zoladex Implant®); goserelin acetate (Zoladex®); histrelin acetate (Histrelin implant®); hydroxyurea (Hydrea®); Ibritumomab Tiuxetan (Zevalin®); idarubicin (Idamycin®); ifosfamide (IFEX®); imatinib mesylate (Gleevec®); interferon alfa 2a (Roferon A®); Interferon alfa-2b (Intron A®); iobenguane I 123 injection (AdreView®); irinotecan (Camptosar®); ixabepilone (Ixempra®); lapatinib tablets (Tykerb®); lenalidomide (Revlimid®); letrozole (Femara®); leucovorin (Wellcovorin®, Leucovorin®); Leuprolide Acetate (Eligard®); levamisole (Ergamisol®); lomustine, CCNU (CeeBU®); meclorethamine, nitrogen mustard (Mustargen®); megestrol acetate (Megace®); melphalan, L- PAM (Alkeran®); mercaptopurine, 6-MP (Purinethol®); mesna (Mesnex®); mesna (Mesnex tabs®); methotrexate (Methotrexate®); methoxsalen (Uvadex®); 8-methoxypsoralen; mitomycin C (Mutamycin®); mitotane (Lysodren®); mitoxantrone (Novantrone®); mitramycin; nandrolone SIR-ONC-00001 phenpropionate (Durabolin-50®); nelarabine (Arranon®); nilotinib (Tasigna®); Nofetumomab (Verluma®); ofatumumab (Arzerra®); Oprelvekin (Neumega®); oxaliplatin (Eloxatin®); paclitaxel (Paxene®); paclitaxel (Taxol®); paclitaxel protein-bound particles (Abraxane®); palifermin (Kepivance®); pamidronate (Aredia®); panitumumab (Vectibix®); pazopanib tablets (Votrienttm®); pegademase (Adagen (Pegademase Bovine)®); pegaspargase (Oncaspar®); Pegfilgrastim (Neulasta®); pemetrexed disodium (Alimta®); pentostatin (Nipent®); pipobroman (Vercyte®); plerixafor (Mozobil®); plicamycin, mithramycin (Mithracin®); porfimer sodium (Photofrin®); pralatrexate injection (Folotyn®); procarbazine (Matulane®); quinacrine (Atabrine®); rapamycin; Rasburicase (Elitek®); raloxifene hydrochloride (Evista®); Rituximab (Rituxan®); romidepsin (Istodax®); romiplostim (Nplate®); sargramostim (Leukine®); Sargramostim (Prokine®); sorafenib (Nexavar®); streptozocin (Zanosar®); sunitinib maleate (Sutent®); talc (Sclerosol®); tamoxifen (Nolvadex®); temozolomide (Temodar®); temsirolimus (Torisel®); teniposide, VM-26 (Vumon®); testolactone (Teslac®); thioguanine, 6-TG (Thioguanine®); thiopurine; thiotepa (Thioplex®); topotecan (Hycamtin®); toremifene (Fareston®); Tositumomab (Bexxar®); Tositumomab/I-131 tositumomab (Bexxar®); trans- retinoic acid; Trastuzumab (Herceptin®); tretinoin, ATRA (Vesanoid®); triethylenemelamine; Uracil Mustard (Uracil Mustard Capsules®); valrubicin (Valstar®); vinblastine (Velban®); vincristine (Oncovin®); vinorelbine (Navelbine®); vorinostat (Zolinza®); wortmannin; and zoledronate (Zometa®).
[0282] The invention also provides a combination comprising an siNA molecule of the invention comprising at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A) and/or a pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof together with another MET inhibitor.
[0283] The combinations referred to above can conveniently be presented for use in the form of a pharmaceutical formulation and thus pharmaceutical compositions comprising a combination as defined above together with a pharmaceutically acceptable diluent or carrier represent a further aspect of the invention.
[0284] The individual compounds of such combinations can be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations. In one embodiment, the individual compounds will be administered simultaneously in a combined pharmaceutical formulation. SIR-ONC-00001
[0285] Thus, the described molecules could be used in combination with one or more known compounds, treatments, or procedures to prevent or treat diseases, disorders, conditions, and traits described herein in a subject or organism as are known in the art, such as other MET inhibitors.
3. Therapeutic Applications
[0286] The present body of knowledge in MET research indicates the need for methods that can regulate MET expression for therapeutic use.
[0287] Thus, one aspect of the invention comprises a method of treating a subject including, but not limited to, a human suffering from a condition which is mediated by the action, or by loss of action, of MET gene expression, which method comprises administering to said subject an effective amount of a double- stranded siNA molecule of the invention. In one embodiment of this aspect, the siNA molecules comprises at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A). In another embodiment of this aspect, the condition is or is caused by cancer. Thus, in certain embodiments the molecules and compositions of the instant invention are useful in a method for treating cancer. Cancers treatable according to this aspect of the invention Cardiac: sarcoma fibrosarcoma, rhabdomyosarcoma, and liposarcoma; Lung: bronchogenic carcinoma, squamous cell carcinoma, undifferentiated small cell carcinoma, undifferentiated large cell carcinoma, lung adenocarcinoma, alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous harmartoma, mesothelioma, non-small cell lung carcinoma, small cell lung cancer, and lung carcinoma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma), stomach (carcinoma, leiomyosarcoma), pancreatic cancer (ductal adenocarcinoma, gastrinoma), rectal cancer, colorectal carcinoma, colon cancer, gastric cancer, squamous cell carcinoma, gastric carcinoma, gastric adenocarcinomas, gastrinoma, and gall bladder carcinoma; Genitourinary tract: kidney carcinoma, bladder carcinoma, urethra, prostate (carcinoma, sarcoma, leiomyosarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma) clear cell carcinoma, papillary renal cell carcinoma, SIR-ONC-00001
Wilms' Tumor, and Cholangiocarcinoma; Liver: hepatoma, hepatocellular carcinoma, cholangiocarcinoma, hepatoblastoma, and liver carcinoma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, multiple myeloma, malignant giant cell tumor benign chondroma, giant cell tumors, osteosarcoma (OS), chordoma, Synovial Sarcoma, and Clear-cell sarcoma; Nervous system: meninges (meningioma, gliomatosis), brain (astrocytoma, glioblastoma), spinal cord (neurofibroma, meningioma, glioma), neuroblastoma, meningioma, neurofibroma, schwannoma, and esophogeal carcinoma; Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, cervical adenocarcinoma, pre-tumor cervical dysplasia), and ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma]); Hematologic: Adult T Cell Leukemia, Lymphomas, Multiple Myeloma, Diffuse large B-cell lymphoma, histiocytic lymphoma, Acute Myelogenous Leukemia, and Chronic Myeloid Leukemia; Skin: melanoma (malignant), basal cell carcinoma, Kaposi's sarcoma, head and neck squamous cell carcinomas, head and neck carcinoma; Breast: breast carcinoma and ductal adenocarcinoma; Thyroid: Nasopharyngeal carcinoma and thyroid carcinoma; Musculoskeletal sarcomas: Osteosarcoma, Rhabdomyosarcoma, and Synovial Sarcoma; Soft tissue sarcomas: Kaposi's Sarcoma, Leimyosarcoma, and MFH/Fibrosarcoma; Metastasis: ovarian cancer, childhood hepatocellular carcinoma, metastatic head and neck squamous cell carcinomas, gastric cancers, breast cancer, colorectal cancer, cervical cancer, lung cancer, nasopharyngeal cancer, and glioblastoma and sarcomas.
[0288] In one embodiment, the siNA molecules of the instant invention are useful in a method for treating or preventing cancer selected from: head and neck squamous cell carcinomas, histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, non- small cell lung cancer, pancreatic cancer, colorectal cancer, cervical cancer, liver cancers, gastric cancers, colon cancers, glioblastomas, thyroid cancers and breast carcinoma. In yet another embodiment, the compounds of the instant invention are useful in a method for treating or preventing cancer selected from: histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, pancreatic cancer, liver cancers, gastric cancers, colon cancers, multiple myeloma, glioblastomas and breast carcinoma. In still another embodiment, the compounds of the instant invention are useful for treating liver cancer, such as hepatoma, hepatocellular carcinoma, cholangiocarcinoma, hepatoblastoma, and liver carcinoma. SIR-ONC-00001
[0289] In another embodiment, the siNA molecules of the instant invention are useful in a method for the prevention or modulation of the metastases of cancer cells and cancer. In particular, the siNA molecules of the instant invention are useful in a method to prevent or modulate the metastases of ovarian cancer, childhood hepatocellular carcinoma, metastatic head and neck squamous cell carcinomas, gastric cancers, breast cancer, colorectal cancer, cervical cancer, lung cancer, nasopharyngeal cancer, pancreatic cancer, glioblastoma and sarcomas.
[0290] In certain embodiments, the administration of the siNA molecule is via local administration or systemic administration. In other embodiments, the invention features contacting the subject or organism with an siNA molecule of the invention via local administration to relevant tissues or cells, such as lung cells and tissues, such as via pulmonary delivery. In yet other embodiments, the invention features contacting the subject or organism with an siNA molecule of the invention via systemic administration (such as via intravenous or subcutaneous administration of siNA) to relevant tissues or cells, such as cancerous tissues or cells in a subject or organism.
[0291] siNA molecules of the invention are also used as reagents in ex vivo applications. For example, siNA reagents are introduced into tissue or cells that are transplanted into a subject for therapeutic effect. The cells and/or tissue can be derived from an organism or subject that later receives the explant, or can be derived from another organism or subject prior to transplantation. The siNA molecules can be used to modulate the expression of one or more genes in the cells or tissue, such that the cells or tissue obtain a desired phenotype or are able to perform a function when transplanted in vivo. In one embodiment, certain MET target cells from a patient are extracted. These extracted cells are contacted with MET siNAs targeting a specific nucleotide sequence within the cells under conditions suitable for uptake of the siNAs by these cells {e.g., using delivery reagents such as cationic lipids, liposomes and the like or using techniques such as electroporation to facilitate the delivery of siNAs into cells). The cells are then reintroduced back into the same patient or other patients.
[0292] For therapeutic applications, a pharmaceutically effective dose of the siNA molecules or pharmaceutical compositions of the invention is administered to the subject. A pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat SIR-ONC-00001
(alleviate a symptom to some extent, preferably all of the symptoms) of a disease state. One skilled in the art can readily determine a therapeutically effective dose of the siNA of the invention to be administer to a given subject, by taking into account factors, such as the size and weight of the subject, the extent of the disease progression or penetration, the age, health, and sex of the subject, the route of administration m and whether the administration is regional or systemic. Generally, an amount between 0.1 mg/kg and 100 mg/kg body weight/day of active ingredients is administered dependent upon potency of the negatively charged polymer. The siNA molecules of the invention can be administered in a single dose or in multiple doses.
[0293] siNA molecules of the instant invention can be administered once monthly, once weekly, once daily (QD), or divided into multiple monthly, weekly, or daily doses, such as, for example, but not limitation, twice daily (BID), three times daily (TID), once every two weeks.
[0294] In addition, the administration can be continuous, i.e., every day, or intermittently. For example, intermittent administration of a compound of the instant invention may be administration one to six days per week or it may mean administration in cycles (e.g. daily administration for two to eight consecutive weeks, then a rest period with no administration for up to one week) or it may mean administration on alternate days.
G. Administration
[0295] Compositions or formulations can be administered in a variety of ways. Non-limiting examples of administration methods of the invention include oral, buccal, sublingual, parenteral (i.e., intraarticularly, intravenously, intraperitoneally, subcutaneously, or intramuscularly), local rectal administration or other local administration. In one embodiment, the composition of the invention can be administered by insufflation and inhalation. Administration can be accomplished via single or divided doses. In some embodiments, the pharmaceutical compositions are administered intravenously or intraperitoneally by a bolus injection (see, e.g., U.S. Pat. No. 5,286,634). Lipid nucleic acid particles can be administered by direct injection at the site of disease or by injection at a site distal from the site of disease (see, e.g., Culver, HUMAN GENE THERAPY, MaryAnn Liebert, Inc., Publishers, New York. pp. 70-71(1994)). In one embodiment, the siNA molecules of the invention and formulations or compositions SIR-ONC-00001 thereof are administered to a cell, subject, or organism as is described herein and as is generally known in the art.
1. In Vivo Administration
[0296] In any of the methods of treatment of the invention, the siNA can be administered to the subject systemically as described herein or otherwise known in the art, either alone as a monotherapy or in combination with additional therapies described herein or as are known in the art. Systemic administration can include, for example, pulmonary (inhalation, nebulization etc.) intravenous, subcutaneous, intramuscular, catheterization, nasopharangeal, transdermal, or oral/gastrointestinal administration as is generally known in the art.
[0297] In any of the methods of treatment or prevention of the invention, the siNA can be administered to the subject locally or to local tissues as described herein or otherwise known in the art, either alone as a monotherapy or in combination with additional therapies as are known in the art. Local administration can include, for example, inhalation, nebulization, catheterization, implantation, direct injection, dermal/transdermal application, patches, stenting, ear/eye drops, or portal vein administration to relevant tissues, or any other local administration technique, method or procedure, as is generally known in the art.
[0298] In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered to the liver as is generally known in the art (see for example Wen et al, 2004, World J Gastroenterol., 10, 244-9; Murao et ah, 2002, Pharm Res., 19, 1808-14; Liu et al, 2003, gene Ther., 10, 180-7; Hong et al, 2003, / Pharm Pharmacol, 54, 51-8; Herrmann et al., 2004, Arch Virol., 149, 1611-7; and Matsuno et al., 2003, gene Ther., 10, 1559-66).
[0299] In one embodiment, the invention features the use of methods to deliver the siNA molecules of the instant invention to hematopoietic cells, including monocytes and lymphocytes. These methods are described in detail by Hartmann et al., 1998, J. Phamacol. Exp. Ther., 285(2), 920-928; Kronenwett et al, 1998, Blood, 91(3), 852-862; Filion and Phillips, 1997, Biochim. Biophys. Acta., 1329(2), 345-356; Ma and Wei, 1996, Leuk. Res., 20(11/12), 925-930; and Bongartz et al, 1994, Nucleic Acids Research, 22(22), 4681-8. SIR-ONC-00001
[0300] In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered directly or topically (e.g., locally) to the dermis or follicles as is generally known in the art (see for example Brand, 2001, Curr. Opin. Mol. Ther., 3, 244-8; Regnier et al., 1998, J. Drug Target, 5, 275-89; Kanikkannan, 2002, BioDrugs, 16, 339-47; Wraight et al, 2001, Pharmacol. Ther., 90, 89-104; and Preat and Dujardin, 2001, STP PharmaSciences, 11, 57-68). In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered directly or topically using a hydroalcoholic gel formulation comprising an alcohol (e.g., ethanol or isopropanol), water, and optionally including additional agents such isopropyl myristate and carbomer 980. In other embodiments, the siNA are formulated to be administered topically to the nasal cavity.. Topical preparations can be administered by one or more applications per day to the affected area; over skin areas occlusive dressings can advantageously be used. Continuous or prolonged delivery can be achieved by an adhesive reservoir system.
[0301] In one embodiment, an siNA molecule of the invention is administered iontophoretically, for example to a particular organ or compartment (e.g., the eye, back of the eye, heart, liver, kidney, bladder, prostate, tumor, CNS etc.). Non-limiting examples of iontophoretic delivery are described in, for example, WO 03/043689 and WO 03/030989, which are incorporated by reference in their entireties herein.
[0302] In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered to the lung as is described herein and as is generally known in the art. In another embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered to lung tissues and cells as is described in U.S. Patent Publication Nos. 2006/0062758; 2006/0014289; and 2004/0077540.
2. Aerosols and Delivery Devices a. Aerosol Formulations
[0303] The compositions of the present invention, either alone or in combination with other suitable components, can be made into aerosol formulations (i.e., they can be "nebulized") to be administered via inhalation (e.g., intranasally or intratracheally) (see, Brigham et al., Am. J. Set, SIR-ONC-00001
298:278 (1989)). Aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.
[0304] In one embodiment, the siNA molecules of the invention and formulations thereof are administered via pulmonary delivery, such as by inhalation of an aerosol or spray dried formulation administered by an inhalation device or nebulizer, providing rapid local uptake of the nucleic acid molecules into relevant pulmonary tissues. Solid particulate compositions containing respirable dry particles of micronized nucleic acid compositions can be prepared by grinding dried or lyophilized nucleic acid compositions, and then passing the micronized composition through, for example, a 400 mesh screen to break up or separate out large agglomerates. A solid particulate composition comprising the siNA compositions of the invention can optionally contain a dispersant which serves to facilitate the formation of an aerosol as well as other therapeutic compounds. A suitable dispersant is lactose, which can be blended with the nucleic acid compound in any suitable ratio, such as a 1 to 1 ratio by weight.
[0305] Spray compositions comprising siNA molecules or compositions of the invention can, for example, be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurized packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant. In one embodiment, aerosol compositions of the invention suitable for inhalation can be either a suspension or a solution and generally contain an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A), and a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1, 2,3,3, 3-heptafluoro-n-propane or a mixture thereof. The aerosol composition can optionally contain additional formulation excipients well known in the art such as surfactants. Non-limiting examples include oleic acid, lecithin or an oligolactic acid or derivative such as those described in W094/21229 and W098/34596 and co-solvents for example ethanol. In one embodiment a pharmaceutical aerosol formulation of the invention comprising a compound of the invention and a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof as propellant, optionally in combination with a surfactant and/or a co- solvent. SIR-ONC-00001
[0306] The aerosol formulations of the invention can be buffered by the addition of suitable buffering agents.
[0307] Aerosol formulations can include optional additives including preservatives if the formulation is not prepared sterile. Non-limiting examples include, methyl hydroxybenzoate, anti-oxidants, flavorings, volatile oils, buffering agents and emulsifiers and other formulation surfactants. In one embodiment, fluorocarbon or perfluorocarbon carriers are used to reduce degradation and provide safer biocompatible non-liquid particulate suspension compositions of the invention (e.g., siNA and/or LNP formulations thereof). In another embodiment, a device comprising a nebulizer delivers a composition of the invention (e.g., siNA and/or LNP formulations thereof) comprising fluorochemicals that are bacteriostatic thereby decreasing the potential for microbial growth in compatible devices.
[0308] Capsules and cartridges comprising the composition of the invention for use in an inhaler or insufflator, of for example gelatine, can be formulated containing a powder mix for inhalation of a compound of the invention and a suitable powder base such as lactose or starch. In one embodiment, each capsule or cartridge contains an siNA molecule comprising at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101 ; or formula (A), and one or more excipients. In another embodiment, the compound of the invention can be presented without excipients such as lactose
[0309] The aerosol compositions of the present invention can be administered into the respiratory system as a formulation including particles of respirable size, e.g. particles of a size sufficiently small to pass through the nose, mouth and larynx upon inhalation and through the bronchi and alveoli of the lungs. In general, respirable particles range from about 0.5 to 10 microns in size. In one embodiment, the particulate range can be from 1 to 5 microns. In another embodiment, the particulate range can be from 2 to 3 microns. Particles of non- respirable size which are included in the aerosol tend to deposit in the throat and be swallowed, and the quantity of non-respirable particles in the aerosol is thus minimized. For nasal administration, a particle size in the range of 10-500 um is preferred to ensure retention in the nasal cavity. SIR-ONC-00001
[0310] In some embodiments, an siNA composition of the invention is administered topically to the nose for example, for the treatment of rhinitis, via pressurized aerosol formulations, aqueous formulations administered to the nose by pressurized pump or by nebulization. Suitable formulations contain water as the diluent or carrier for this purpose. In certain embodiments, the aqueous formulations for administration of the composition of the invention to the lung or nose can be provided with conventional excipients such as buffering agents, tonicity modifying agents and the like. b. Devices
[0311] The siNA molecules of the invention can be formulated and delivered as particles and/or aerosols as discussed above and dispensed from various aerosolization devices known by those of skill in the art.
[0312] Aerosols of liquid or non-liquid particles comprising an siNA molecule or formulation of the invention can be produced by any suitable means, such as with a device comprising a nebulizer (see for example US 4,501,729) such as ultrasonic or air jet nebulizers.
[0313] Solid particle aerosols comprising an siNA molecule or formulation of the invention and surfactant can be produced with any solid particulate aerosol generator. One type of solid particle aerosol generator used with the siNA molecules of the invention is an insufflator. A second type of illustrative aerosol generator comprises a metered dose inhaler ("MDI"). MDIs containing siNA molecules or formulations taught herein can be prepared by methods of the art (for example, see Byron, above and WO96/32099).
[0314] The siNA molecules can also be formulated as a fluid formulation for delivery from a fluid dispenser, such as those described and illustrated in WO05/044354.
[0315] In certain embodiments of the invention, nebulizer devices are used in applications for conscious, spontaneously breathing subjects, and for controlled ventilated subjects of all ages. The nebulizer devices can be used for targeted topical and systemic drug delivery to the lung. In one embodiment, a device comprising a nebulizer is used to deliver an siNA molecule or formulation of the invention locally to lung or pulmonary tissues. In another embodiment, a SIR-ONC-00001 device comprising a nebulizer is used to deliver a an siNA molecule or formulation of the invention systemically.
H. Other Applications/Uses of siNA Molecules of the Invention
[0316] The siNA molecules of the invention can also be used for diagnostic applications, research applications, and/or manufacture of medicants.
[0317] In one aspect, the invention features a method for diagnosing a disease, trait, or condition in a subject comprising administering to the subject a composition of the invention under conditions suitable for the diagnosis of the disease, trait, or condition in the subject.
[0318] In one embodiment, siNA molecules of the invention are used to down regulate or inhibit the expression of MET proteins arising from haplotype polymorphisms that are associated with a trait, disease or condition in a subject or organism. Analysis of MET genes, or MET protein or RNA levels can be used to identify subjects with such polymorphisms or those subjects who are at risk of developing traits, conditions, or diseases described herein. These subjects are amenable to treatment, for example, treatment with siNA molecules of the invention and any other composition useful in treating diseases related to target gene expression. As such, analysis of MET protein or RNA levels can be used to determine treatment type and the course of therapy in treating a subject. Monitoring of MET protein or RNA levels can be used to predict treatment outcome and to determine the efficacy of compounds and compositions that modulate the level and/or activity of certain MET proteins associated with a trait, disorder, condition, or disease.
[0319] In another embodiment, the invention comprises use of a double-stranded nucleic acid according to the invention for use in the manufacture of a medicament. In an embodiment, the medicament is for use in treating a condition that is mediated by the action, or by loss of action, of MET. In one embodiment, the medicament is for use for the treatment of cancer. In an embodiment, the medicament is for use for the treatment of head and neck squamous cell carcinomas, histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, non- small cell lung cancer, pancreatic cancer, papillary renal cell carcinoma, liver cancer, gastric cancer, colon cancer, multiple myeloma, glioblastomas and breast carcinoma. In a particular embodiment, the SIR-ONC-00001 use is for the treatment of liver cancer selected from the group consisting hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma, carcinoma.
[0320] In certain embodiments, siNAs wherein at least one strand comprises at least a 15 nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 1049, SEQ ID NO: 43, SEQ ID NO: 1091, SEQ ID NO: 51, SEQ ID NO: 1099, SEQ ID NO: 53, or SEQ ID NO: 1101; or formula (A),are for use in a method for treating a cancer, such as, for example but not limitation, of head and neck squamous cell carcinomas, histiocytic lymphoma, lung adenocarcinoma, small cell lung cancer, non-small cell lung cancer, pancreatic cancer, papillary renal cell carcinoma, liver cancer, gastric cancer, colon cancer, multiple myeloma, glioblastomas and breast carcinoma.
I. Examples
[0321] The invention will now be illustrated with the following non-limiting examples. Those of skill in the art will readily recognize a variety of non-critical parameters which can be changed or modified to yield essentially the same results.
Example 1: Design, Synthesis, and Identification of siNAs Active Against MET. MET siNA Synthesis
[0322] A series of siNA molecules were designed, synthesized and evaluated for efficacy against MET gene expression. Certain MET sequences were designed and selected by methods set forth in U.S. Application No. 60/182,605. Other sequences were designed and selected using a proprietary algorithm. The primary criteria for design of certain of the MET sequences for human siNAs were (i) homology between two species (human and rhesus monkey) and (ii) high efficacy scores as determined by a proprietary algorithm. The effects of the siNAs on MET RNA levels. The target sequences of the siNAs that were selected are set forth in Table la (target sequences). The sense and antisense strands of the siNA sequences corresponding to the target sequences in Table la are set forth in Table lb. Various chemically modified siNAs that were synthesized are set forth in Table lc.
Table la: MET Target Sequences, noting target sites. SIR-ONC-00001
Figure imgf000093_0001
SIR-ONC-00001
Figure imgf000094_0001
SIR-ONC-00001
Figure imgf000095_0001
SIR-ONC-00001
Figure imgf000096_0001
SIR-ONC-00001
Figure imgf000097_0001
SIR-ONC-00001
Figure imgf000098_0001
SIR-ONC-00001
Figure imgf000099_0001
Table lb. Various c-MET siNA sequences corresponding to the identified target sequence.
Figure imgf000099_0002
SIR-ONC-00001
Figure imgf000100_0001
SIR-ONC-00001
Figure imgf000101_0001
SIR-ONC-00001
Figure imgf000102_0001
SIR-ONC-00001
Figure imgf000103_0001
SIR-ONC-00001
Figure imgf000104_0001
SIR-ONC-00001
Figure imgf000105_0001
[0323] For each oligonucleotide of a target sequence, the two individual, complementary strands of the siNA were synthesized separately using solid phase synthesis, then purified separately by reversed phase solid phase extraction (SPE). The complementary strands were annealed to form the double strand (duplex) and delivered in the desired concentration and buffer of choice.
[0324] Briefly, the single strand oligonucleotides were synthesized using phosphoramidite chemistry on an automated solid-phase synthesizer, using procedures as are generally known in the art (see for example U.S. Application No. 12/064,014). A synthesis column was packed with solid support derivatized with the first nucleoside residue (natural or chemically modified). Synthesis was initiated by detritylation of the acid labile 5'-0-dimethoxytrityl group to release the 5'-hydroxyl. A suitably protected phosphoramidite and a suitable activator in acetonitrile were delivered simultaneously to the synthesis column resulting in coupling of the amidite to the SIR-ONC-00001
5'-hydroxyl. The column was then washed with a solvent, such as acetonitrile. An oxidizing solution, such as an iodine solution was pumped through the column to oxidize the phosphite triester linkage P(III) to its phosphotriester P(V) analog. Unreacted 5'-hydroxyl groups were capped using reagents such as acetic anhydride in the presence of 2,6-lutidine and N- methylimidazole. The elongation cycle was resumed with the detritylation step for the next phosphoramidite incorporation. This process was repeated until the desired sequence was synthesized. The synthesis concluded with the final 5 '-terminus protecting group (trityl or 5'-0- dimethoxytrityl) .
[0325] Upon completion of the synthesis, the solid-support and associated oligonucleotide were dried under argon pressure or vacuum. Aqueous base was added and the mixture was heated to effect cleavage of the succinyl linkage, removal of the cyanoethyl phosphate protecting group, and deprotection of the exocyclic amine protection.
[0326] The following process was performed on single strands that do not contain ribonucleotides. After treating the solid support with the aqueous base, the mixture was filtered to separate the solid support from the deprotected crude synthesis material. The solid support was then rinsed with water, which is combined with the filtrate. The resultant basic solution allows for retention of the 5'-0-dimethoxytrityl group to remain on the 5' terminal position (trityl- on).
[0327] For single strands that contain ribonucleotides, the following process was performed. After treating the solid support with the aqueous base, the mixture was filtered to separate the solid support from the deprotected crude synthesis material. The solid support was then rinsed with dimethylsulfoxide (DMSO), which was combined with the filtrate. Fluoride reagent, such as triethylamine trihydrofluoride, was added to the mixture, and the solution was heated. The reaction was quenched with suitable buffer to provide a solution of crude single strand with the 5'-0-dimethoxytrityl group on the final 5' terminal position.
[0328] The trityl-on solution of each crude single strand was purified using chromatographic purification, such as SPE RPC purification. The hydrophobic nature of the trityl group permits stronger retention of the desired full-length oligo than the non-tritylated truncated failure sequences. The failure sequences were selectively washed from the resin with a suitable solvent, SIR-ONC-00001 such as low percent acetonitrile. Retained oligonucleotides were then detritylated on-column with trifluoroacetic acid to remove the acid-labile trityl group. Residual acid was washed from the column, a salt exchange was performed, and a final desalting of the material commenced . The full-length oligo was recovered in a purified form with an aqueous-organic solvent. The final product was then analyzed for purity (HPLC), identity (Maldi-TOF MS), and yield (UV A26o)- The oligos were dried via lyophilization or vacuum condensation.
[0329] Annealing: Based on the analysis of the product, the dried oligos were dissolved in appropriate buffers followed by mixing equal molar amounts (calculated using the theoretical extinction coefficient) of the sense and antisense oligonucleotide strands. The solution was then analyzed for purity of duplex by chromatographic methods and desired final concentration. If the analysis indicated an excess of either strand, then the additional non-excess strand was titrated until duplexing was complete. When analysis indicated that the target product purity has been achieved the material was delivered and ready for use.
[0330] Below is a table showing various modified siNAs synthesized using this protocol.
Table lc: MET siNA Strands Synthesized (Antisense sequences are readily identified as being complementary to the target sequence shown).
Figure imgf000107_0001
SIR-ONC-00001
Figure imgf000108_0001
SIR-ONC-00001
Figure imgf000109_0001
SIR-ONC-00001
Figure imgf000110_0001
SIR-ONC-00001
Figure imgf000111_0001
SIR-ONC-00001
Figure imgf000112_0001
SIR-ONC-00001
Figure imgf000113_0001
SIR-ONC-00001
Figure imgf000114_0001
SIR-ONC-00001
Figure imgf000115_0001
SIR-ONC-00001
Figure imgf000116_0001
SIR-ONC-00001
Figure imgf000117_0001
SIR-ONC-00001
Figure imgf000118_0001
SIR-ONC-00001
Figure imgf000119_0001
SIR-ONC-00001
Figure imgf000120_0001
SIR-ONC-00001
Figure imgf000121_0001
SIR-ONC-00001
Figure imgf000122_0001
SIR-ONC-00001
Figure imgf000123_0001
SIR-ONC-00001
Figure imgf000124_0001
SIR-ONC-00001
Figure imgf000125_0001
wherein:
A, C, G, and U = ribose A, C, G or U
c and u = 2'-deoxy-2'-fhioro C or U
A, U and G = 2'-0-methyl (2'-OMe) A U or G
A and G = deoxy A or G
B = inverted abasic
T = thymidine
s = phosphorothioate linkage
Further Synthesis Steps for Commercial Preparation
[0331] Once analysis indicates that the target product purity has been achieved after the annealing step, the material is transferred to the tangential flow filtration (TFF) system for concentration and desalting, as opposed to doing this prior to the annealing step.
[0332] Ultrafiltration: The annealed product solution is concentrated using a TFF system containing an appropriate molecular weight cut-off membrane. Following concentration, the product solution is desalted via diafiltration using Milli-Q water until the conductivity of the filtrate is that of water.
[0333] Lyophilization: The concentrated solution is transferred to a bottle, flash frozen and attached to a lyophilizer. The product is then freeze-dried to a powder. The bottle is removed from the lyophilizer and is now ready for use.
Initial Screening Protocol (96-Well Plate Transfections)
Cell Culture Preparation:
[0334] Human hepatoma cell line, HepG2, rhesus kidney epithelial cell line, LLC-MK2 Derivative, and cynomulgus skin fibroblast cell line, Cynom-Kl, were grown in modified Eagle's SIR-ONC-00001 medium. All the culture media were supplemented with 10% fetal bovine serum, 100μg/mL streptomycin, lOOU/mL penicillin, and 1% sodium bicarbonate.
Transfection and Screening
[0335] Cells were plated in all wells of tissue-culture treated, 96-well plates at a final count of 3500 (HepG2 and LLC-MK2 Derivative) or 5000 (Cynom-Kl) cells/well in ΙΟΟμΓ-. of the appropriate culture media. For duration assays, HepG2 cells were plated at 1200 cell/swell in ΙΟΟμί medium. The cells were cultured for overnight after plating at 37°C in the presence of 5% C02.
[0336] On the next day, complexes containing siNA and RNAiMax (Invitrogen) were created as follows. A solution of RNAiMax diluted 33-fold in OPTI-MEM was prepared. In parallel, solutions of the siNAs for testing were prepared to a final concentration of 120 nM in OPTI-MEM. After incubation of RNAiMax/OPTI-MEM solution at room temperature for 5 min, an equal volume of the siNA solution and the RNAiMax solution were added together for each of the siNAs.
[0337] Mixing resulted in a solution of siNA/RNAiMax where the concentration of siNA was 60 nM. This solution was incubated at room temperature for 20 minutes. After incubation, 20 μΐ^ of the solution was added to each of the relevant wells. The final concentration of siNA in each well was 10 nM and the final volume of RNAiMax in each well was 0.3ul.
[0338] For low concentration screens, siNAs were transfected at 200, 150, 100 or 75pM per well. For 12-point dose response curve studies, the siNA series are 6-fold serial dilution starting at 30nM or 4-fold serial dilution starting at 40nM. All transfections were set up as multiple biological replicates.
[0339] The time of incubation with the RNAiMax -siNA complexes was 24 hours and there was no change in media between transfection and harvesting for screening and dose response curve studies. For duration assays, the time of incubation with the RNAiMax -siNA complexes was 24, 72, and 120 hours. There was no change in media between transfection and harvesting for 24- and 72-hour time points. Media was replaced with fresh media 72 hours after transfection for 120-hour time point.. SIR-ONC-00001
Cells-to-Ct and reverse transcription reactions
[0340] The culture medium was aspirated and discarded from the wells of the culture plates at the desired time points. The transfected cells were washed once with 50uL DPBS solution per well. Fifty microliters per well of the Lysis Solution from the TaqMan® Gene Expression Cells- to-CT™ Kit (Applied Biosystems, Cat# 4399002) supplemented with DNase I was added directly to the plates to lyse the cells. Five microliters per well of Stop Solution from the same kit was added to the plates to inactivate DNase I 5 min. later. The lysis plates were incubated for at least 2 min. at room temperature. The plates can be stored for 2 hours at 4°C, or -80 °C for two months.
[0341] Each well of the reverse transcription plate required lOuL of 2X reverse transcriptase buffer, luL of 20X reverse transcription enzyme and 2uL of nuclease-free water. The reverse transcription master mix was prepared by mixing 2X reverse transcription buffer, 20X reverse transcription enzyme mix, and nuclease-free water. 13uL of the reverse transcription master mix was dispensed into each well of the reverse transcription plate (semi-skirted). A separate reverse transcription plate was prepared for each cell plate. A separate reverse transcription plate was prepared for each cell plate. Seven microliters per lysate from the cell lysis procedure described above was added into each well of the reverse transcription plate. The plate was sealed and spun on a centrifuge (lOOOrpm for 30 seconds) to settle the contents to the bottom of the reverse transcription plate. The plate was placed in a thermocycler at 37 °C for 60 min, 95 °C for 5 min, and 4 °C until the plate is removed from the thermocycler. Upon removal, if not used immediately, the plate was frozen at -20 °C.
[0342] For duration assays, a similar protocol was followed, however, the cells were lysed 1, 3, and 5 days after transfection. cDNA was generated using Cells-to-Ct™ Kit (Applied Biosystems).
Quantitative RT-PCR (Taqman)
[0343] A series of probes and primers were used to detect the various mRNA transcripts of the genes of MET and GAPDH. All Taqman probes and primers for the experiments here-in described were supplied as pre-validated sets by Applied Biosystems, Inc. (see Table 2). SIR-ONC-00001
Table 2: Probes and primers used to carry out Real-Time RT/PCR (Taqman) reactions for MET mRNA analysis.
Figure imgf000128_0001
[0344] The assays were performed on an ABI 7900 instrument, according to the manufacturer's instructions. A TaqMan Gene Expression Master Mix (provided in the Cells-to- CT™ Kit, Applied Biosystems, Cat # 4399002) was used. The PCR reactions were carried out at 50 °C for 2 min, 95 °C for 10 min followed by 40 cycles at 95 °C for 15 sees and 60 °C for 1 min.
[0345] Within each experiment, the baseline was set in the exponential phase of the amplification curve, and based on the intersection point of the baselines with the amplification curve, a Ct value was assigned by the instrument.
Calculations
[0346] The expression level of the gene of interest and % inhibition of gene expression ( KD) was calculated using Comparative Ct method:
ACt = Ct Target - Ct GAPDH
AACt (log2(fold change)) = ACt (Taiget siNA) - ACt (NTc)
Relative expression level = 2~AACt
KD = 100 x (l - 2" Ct)
[0347] The non-targeting control siNA was, unless otherwise indicated, chosen as the value against which to calculate the percent inhibition (knockdown) of gene expression, because it is the most relevant control.
[0348] Additionally, only normalized data, which reflects the general health of the cell and quality of the RNA extraction, was examined. This was done by looking at the level of two SIR-ONC-00001 different mRNAs in the treated cells, the first being the target mRNA and the second being the normalizer mRNA. This allowed for elimination of siNAs that might be potentially toxic to cells rather than solely knocking down the gene of interest. This was done by comparing the Ct for GAPDH in each well relative to the GAPDH Ct for the entire plate.
[0349] All calculations of IC50S were performed using R.2.9.2 software. The data were analyzed using the sigmoidal dose-response (variable slope) equation for simple ligand binding. In all of the calculations of the percent inhibition (knock-down), the calculation was made relative to the normalized level of expression of the gene of interest in the samples treated with the non-targeting control (Ctrl siNA) unless otherwise indicated.
[0350] The level of protein was quantified using the Bio-Rad VersaDoc Imager according to the protocols of that piece of equipment. A pixel count was performed in each lane using an area of identical size. Each sample was then compared to the appropriate control treated sample and converted to a percent of protein remaining compared to control.
[0351] The effects of lead siNAs on MET protein level were compared to the effects of the universal control using a two tail Student's T-test to obtain a P value. P < 0.05 was considered significant.
Results:
[0352] The MET siNAs were designed and synthesized as described previously. Various siNAs were screened in HepG2 cells. The log 2(fold change) in MET gene expression data upon treatment with various modified MET siNAs in human cells is shown in Table 3a and b. Each screen was performed at 24 hrs. Quantitative RT-PCR was used to assess the level of MET mRNA and the data were normalized to the expression level of GAPDH (an ubiquitously expressed 'house-keeping' gene). Each treatment was then normalized against the non-MET targeting control.
Table 3a: Primary screening data in HepG2 Cells (n = 2-4), recorded as log 2(fold change) in
MET gene expression.
Figure imgf000129_0001
SIR-ONC-00001
Figure imgf000130_0001
Table 3b: Primary screening data in HepG2 Cells (n = 2-4), recorded as log 2(fold change) in
MET gene expression.
Figure imgf000130_0002
SIR-ONC-00001
Figure imgf000131_0001
SIR-ONC-00001
Figure imgf000132_0001
[0353] A subset of siNAs from Table 3a and Table 3b having a large log2(fold change) in the primary screen and/or a low IC50 in dose response curves were tested in human HepG2 cells at a lower concentrations to determine the effect of the MET siNAs on the mRNA transcript of SIR-ONC-00001 human MET at varying concentrations. Table 4 summarizes the data. The siNAs were ranked based on log2(fold change) in MET mRNA expression level.
Table 4: Log2(fold change) of various siNAs tested at low concentrations.
Figure imgf000133_0001
[0354] Select high ranking (lower numerical value) siNAs from Table 4 were further analyzed for efficacy and potency in HepG2cells, LLC-MK2 Derivative and Cynom-Kl cells to ensure that these siNAs functioned well in monkey cells for primate studies. The results for SIR-ONC-00001 these siNAs are shown in Table 5. The potency 50 is the calculated siNA transfection concentration that produces 50% target mRNA knockdown.
Table 5: Dose response data for various siNAs in HepG2 -Human hepatoma, LLC-MK2 Derivative - Rhesus monkey kidney epithelial, and Cynom-Kl - Cynomolgus monkey skin fibroblast cell lines. Calculated maximum AACt is determined from the dose response curve.
Figure imgf000134_0001
Example 2: In Vitro Serum Stability Testing of siNAs
[0355] siNAs were reconstituted as 50μΜ to ΙΟΟμΜ stock solution with H20 and added to human serum pre- warmed to 37 °C to a final concentration of 20μg/mL. The mixture was then incubated at 37°C for 0, 1 and 2 hours. At the end of each time point, the reactions were stopped by mixing with equal volume of Phenomenex Lysis-Loading Buffer. Oligonucleotides were purified in 96-well format by Phenomenex Solid Phase Extraction and lyophilized until dry with Labconco Triad Lyo-00417. The lyophilized samples were reconstituted in 150 μΐ^ of 1 mM EDTA prepared with RNase-free H20. The sample solutions were then diluted 5 fold with 1 mM EDTA for liquid chromatography/mass spectrometry (LC/MS) analysis on Thermo Fisher Orbitrap. Serum metabolites of the siNAs were determined based on the measured molecular weights.
[0356] Results
[0357] In an effort to further optimize and assess the impact of various modifications on siNAs, a set of five target sites were selected to assess further modifications to siNAs targeting these sites. The sites were selected based on demonstrated high knockdown by previously modified siNAs in primary and/or low concentration screen. siNAs targeting these target sites were synthesized with a series of additional modification motifs and tested in a primary screen to SIR-ONC-00001 determine knockdown. Select compounds from the primary screen (those demonstrating a large log2(fold change)) were then tested in low concentration screens and/or in a human serum stability assays. Those siNAs tested in the human serum stability assays were tested to determine the percent of the full-length sense and antisense siNA strands remaining after the time periods of 1 hour and 2 hours. The log2(fold change) data for these siNAs are shown in Table 6. For each target site, the four compounds that had the best overall knockdown (i.e., largest log2(fold change) from the primary and low concentration assays combined were selected along with a control siNA having Stab7/Stab35G chemistry for testing with regard to human serum stability at 1 and 2 hour intervals.
Table 6. Log2(fold change) and serum stability for various modified siNAs including data for low concentration screens.
Figure imgf000135_0001
SIR-ONC-00001
Figure imgf000136_0001
Example 3: In Vitro Duration Assay
[0358] To further assess the effect of various modifications on the siNAs, additional siNAs to the preferred four of the five targets sites shown in Table 6, but with new modification motifs were synthesized as described above and the log2(fold change) (reported as AACt) after 1, 3, and 5 days was determined and compared with a number of modified siNAs previously tested. The results are shown in Table 7. The Rank of the siNA in the primary screen is a ranking of siNAs that have the same target site.
Table 7. Log2(fold change) data and duration data for various modified siNAs.
Figure imgf000136_0002
SIR-ONC-00001
Figure imgf000137_0001
SIR-ONC-00001
Figure imgf000138_0001
Midscale experiments
[0359] Based on an analysis of the results shown in the previous tables several lead siNAs targeting the four target sites used in Table 7 were selected for scaleup and further optimization. Certain of these lead siNAs were further modified in an effort to enhance or reduce certain characteristics, e.g., duration, immunostimulation, etc. For example, it is has been demonstrated that replacement of the TT nucleotide overhang with an 2x O-methyl uridine overhang increases duration.. These compounds are set forth in Table 8 with potency and duration data from compounds that underwent the midscale synthesis and purification. SIR-ONC-00001
Table 8. Log 2 fold change (reported as AACt), potency, and duration data for certain lead siNAs.
Figure imgf000139_0001
Example 3: Testing of Cytokine Induction
[0360] To assess immunostimulative effects of various siNAs of the invention loaded in lipid nanoparticles (DLinDMA/Cholesterol/ S-PEG-C-DMA/DSPC in a 40/48/2/10 ratio), C57B1/6 mice were dosed with a single 3mpk dose of LNP formulated siNAs through tail vein injection. Serum or plasma samples were collected at 3 and 24 hours post-dose. The cytokine and chemokine levels in these samples were measured with the SearchLight IR Cytokine Array from Aushon Biosciences according to the manufacturer's instruction. The cytokines and chemokines measured are IL-la, IL-Ιβ, IL-6, KC, IL-10, IFNy, TNF, GMCSF, ΜΙΡ-Ιβ, MCP-l/JE, and RANTES. The levels of inflammatory cytokines (exp. KC, IFN-g, TNF-a, IL-6) were not induced above the levels of delivery vehicle itself for the siNAs tested, specifically R-
008039841- 001L, R-008282139-000H, R-008282202-000E, R-008282175-000T, and R-
008039842- 001V. SIR-ONC-00001
Example 4: Efficacy Study in TRE-MET Tumor Model
[0361] In vivo efficacy studies were conducted in TRE-MET mice maintained at Taconic Farms. Mice were dosed IV via tail vein injections with LNP encapsulated siNAs or vehicle control using 2 different 3-week dosing schemes: one 1 mg/kg dose for 3 consecutive days or a single 6 mg/kg does per week. In some experiments, the mice were co-dosed with sorafenib at a dose of 100 mg/kg BID every day for 3 weeks. Total tumor burden was measured by micro-CT scan imaging. The animals were sacrificed 5 days after the last siNA dose (Day 23), and normal liver and tumor tissues from each animal were collected for RNA purification. Total RNA was purified using RNeasy 96 kit (Qiagen, Cat# 74182). cDNA was generated from total RNA using High Capacity cDNA Reverse Transcription Kit (Cat#: 4368813). Quantitative PCR reactions were performed with TaqMan Universal PCR Master Mix (Cat#: 4304437). Human MET TaqMan Gene Expression Assay (Hs00179845_ml) and mouse GAPDH TaqMan Gene Expression Assay (Cat#: 4352932E) were used to monitor the mRNA level of both transcripts. The expression level of MET was normalized against GAPDH to minimize technical variations. All reverse transcription and quantitative PCR reagents are from Applied Biosystems, Inc.
[0362] As shown in Table 9 and Figure 11, treatment of TRE-MET mice with various lead siNAs of the invention resulted in tumor growth inhibition achieving up to 80% reduction in tumor growth at 3 weeks and reaching tumor stasis in combination with sorafenib.
Figure imgf000140_0001
SIR-ONC-00001
Figure imgf000141_0001
Example 5: Pharmacodynamic Study in Non-Human Primates
A non-human primate pharmacodynamic study was performed in Rhesus macaque monkeys at New Iberia Research Center of University of Louisiana at Lafayette. The animals were dosed with a single 2.5mpk dose of siNA loaded lipid nanoparticles through intravenous infusion. To monitor target mRNA knockdown, liver biopsies were performed at various time points pre- and post-dose with 16T gauge Menghini needles for about 20mg tissue per animal. Whole blood and serum/plasma were also collected at different time points pre- and post-dose to monitor potential toxicity associated with the treatments. All procedures adhere to the regulations outlined in the USDA Animal Welfare Act (9 CFR, Parts 1, 2 and 3) and the conditions specified in The Guide for Care and Use of Laboratory Animals (ILAR publication, 1996, National Academy Press).
[0363] LNP formulations (DLinDMA/Cholesterol/S-PEG-C-DMA/DSPC in a 40/48/2/10 ratio) comprising several modified siNAs R-008347048-000B, R-008313708-000E, and R- 008313730-000M in an N/P ratio of 6 were tested. Log2(fold change) in MET gene expression was determined on days 3, 7, 14, and 28 days post-dosing. Pre-dose MET expression levels for the monkeys were measured 7 days before the first dosing. As can be seen from Figure 12, the log 2(fold change) is greater than 4 and the percent inhibition of MET gene expression by these siNAs is greater than 90% at day 3. At day 14, this level of greater than 90% inhibition is still maintained by siNA R-008313708-000E.
Example 6: siNAs LNP Formulations
A. General LNP Process Description for DLinDMA Formulations:
[0364] The lipid nanoparticles were prepared by an impinging jet process. The particles were formed by mixing lipids dissolved in alcohol with siNA dissolved in a citrate buffer. The mixing ratio of lipids to siNA is targeted at 45-55% lipid and 65-45% siNA. The lipid solution SIR-ONC-00001 contained a cationic lipid, a helper lipid (cholesterol), PEG (e.g. PEG-C-DMA, PEG-DMG) lipid, and DSPC at a concentration of 5-15 mg/mL with a target of 9-12 mg/mL in an alcohol (for example ethanol). The ratio of the lipids had a mole percent range of 25-98 for the cationic lipid with a target of 35-65, the helper lipid had a mole percent range from 0-75 with a target of 30-50, the PEG lipid has a mole percent range from 1-15 with a target of 1-6, and the DSPC had a mole percent range of 0-15 with a target of 0-12. The siNA solution contained one or more siNA sequences at a concentration range from 0.3 to 1 .0 mg/mL with a target of 0.3 -0.9 mg/mL in a sodium citrate buffered salt solution with pH in the range of 3.5-5. The two liquids were heated to a temperature in the range of 15-40°C, targeting 30-40°C, and then mixed in an impinging jet mixer instantly forming the LNP. The teelD had a range from 0.25 to 1.0 mm and a total flow rate from 10 -600 mL/min. The combination of flow rate and tubing ID had the effect of controlling the particle size of the LNPs between 30 and 200 nm. The solution was then mixed with a buffered solution at a higher pH with a mixing ratio in the range of 1: 1 to 1:3 vol: vol but targeting 1:2 vol: vol. This buffered solution was at a temperature in the range of 15- 40°C, targeting 30-40°C. The mixed LNPs were held from 30 minutes to 2 hrs prior to an anion exchange filtration step. The temperature during incubating was in the range of 15-40°C, targeting 30-40°C. After incubating, the solution was filtered through a 0.8 um filter containing an anion exchange separation step. This process used tubing IDs ranging from 1 mm ID to 5 mm ID and a flow rate from 10 to 2000 mL/min. The LNPs were concentrated and diafiltered via an ultrafiltration process where the alcohol was removed and the citrate buffer was exchanged for the final buffer solution such as phosphate buffered saline. The ultrafiltration process used a tangential flow filtration format (TFF). This process used a membrane nominal molecular weight cutoff range from 30 -500 KD. The membrane format was hollow fiber or flat sheet cassette. The TFF processes with the proper molecular weight cutoff retained the LNP in the retentate and the filtrate or permeate contained the alcohol; citrate buffer; and final buffer wastes. The TFF process is a multiple step process with an initial concentration to a siNA concentration of 1 -3 mg/mL. Following concentration, the LNPs solution was diafiltered against the final buffer for 10 -20 volumes to remove the alcohol and perform buffer exchange. The material was then concentrated an additional 1-3 fold. The final steps of the LNP process were to sterile filter the concentrated LNP solution and vial the product.
Analytical Procedure: SIR-ONC-00001
1) siNA concentration
[0365] The siNA duplex concentrations were determined by Strong Anion-Exchange High- Performance Liquid Chromatography (SAX-HPLC) using Waters 2695 Alliance system (Water Corporation, Milford MA) with a 2996 PDA detector. The LNPs, otherwise referred to as RNAi Delivery Vehicles (RDVs), were treated with 0.5% Triton X-100 to free total siNA and analyzed by SAX separation using a Dionex BioLC DNAPac PA 200 (4 x 250 mm) column with UV detection at 254 nm. Mobile phase was composed of A: 25 mM NaC104, 10 mM Tris, 20% EtOH, pH 7.0 and B: 250 mM NaC104, 10 mM Tris, 20% EtOH, pH 7.0 with a liner gradient from 0-15 min and a flow rate of 1 ml/min. The siNA amount was determined by comparing to the siNA standard curve.
2) Encapsulation rate
[0366] Fluorescence reagent SYBR Gold was employed for RNA quantitation to monitor the encapsulation rate of RDVs. RDVs with or without Triton X-100 were used to determine the free siNA and total siNA amount. The assay is performed using a SpectraMax M5e microplate spectrophotometer from Molecular Devices (Sunnyvale, CA). Samples were excited at 485 nm and fluorescence emission was measured at 530 nm. The siNA amount is determined by comparing to an siNA standard curve.
Encapsulation rate = (1- free siN A/total siNA) xl00%
3) Particle size and polydispersity
[0367] RDVs containing 1 μg siNA were diluted to a final volume of 3 ml with 1 x PBS. The particle size and polydispersity of the samples was measured by a dynamic light scattering method using ZetaPALS instrument (Brookhaven Instruments Corporation, Holtsville, NY). The scattered intensity was measured with He-Ne laser at 25°C with a scattering angle of 90°.
4) Zeta Potential Analysis
[0368] RDVs containing 1 μg siNA were diluted to a final volume of 2 ml with 1 mM Tris buffer (pH 7.4). Electrophoretic mobility of samples was determined using ZetaPALS SIR-ONC-00001 instrument (Brookhaven Instruments Corporation, Holtsville, NY) with electrode and He-Ne laser as a light source. The Smoluchowski limit was assumed in the calculation of zeta potentials.
5) Lipid analysis
[0369] Individual lipid concentrations were determined by Reverse Phase High-
Performance Liquid Chromatography (RP-HPLC) using Waters 2695 Alliance system (Water Corporation, Milford MA) with a Corona charged aerosol detector (CAD) (ESA Biosciences, Inc, Chelmsford, MA). Individual lipids in RDVs were analyzed using an Agilent Zorbax SB- CIS (50 x 4.6 mm, 1.8 μιη particle size) column with CAD at 60°C. The mobile phase was composed of A: 0.1% TFA in H20 and B: 0.1% TFA in IPA. The gradient changed from 60% mobile phase A and 40% mobile phase B from time 0 to 40% mobile phase A and 60% mobile phase B at 1.00 min; 40% mobile phase A and 60% mobile phase B from 1.00 to 5.00 min; 40% mobile phase A and 60% mobile phase B from 5.00 min to 25% mobile phase A and 75% mobile phase B at 10.00 min; 25% mobile phase A and 75% mobile phase B from 10.00 min to 5% mobile phase A and 95% mobile phase B at 15.00 min; and 5% mobile phase A and 95% mobile phase B from 15.00 to 60% mobile phase A and 40% mobile phase B at 20.00 min with a flow rate of 1 ml/min. The individual lipid concentration was determined by comparing to the standard curve with all the lipid components in the RDVs with a quadratic curve fit. The molar percentage of each lipid was calculated based on its molecular weight.
B. General Formulation Procedure for CLinDMA/Cholesterol/PEG-DMG at a ratio of 71.9:20.2:7.9.
[0370] siNA solutions were prepared by dissolving siNAs in 25mM citrate buffer (pH 4.0) at a concentration of 0.8mg/mL. Lipid solutions were prepared by dissolving a mixture of 2S- Octyl-ClinDMA, cholesterol and PEG-DMG at a ratio of 71.9:20.2:7.9 in absolute ethanol at a concentration of about lOmg/mL. Equal volume of siNA and lipid solutions were delivered with two syringe pumps at the same flow rates to a mixing T connector. The resulting milky mixture was collected in a sterile bottle. This mixture was then diluted slowly with an equal volume of citrate buffer, and filtered through a size exclusion hollow fiber cartridge to remove any free siNA in the mixture. Ultra filtration against citrate buffer (pH 4.0) was employed to remove SIR-ONC-00001 ethanol (test stick from ALCO screen), and against PBS (pH 7.4) to exchange buffer. The final LNP was obtained by concentrating to a desired volume and sterile filtered through a 0.2mm filter. The obtained LNPs were characterized in term of particle size, alcohol content, total lipid content, nucleic acid encapsulated, and total nucleic acid concentration.
[0371]
C. General LNP Preparation For Various Formulations in Table 13.
[0372] siNA nanoparticle solutions in Table 13 are prepared by dissolving siNAs and/or carrier molecules in 25 mM citrate buffer (pH 4.0) at a concentration of 0.9 mg/mL. Lipid solutions are prepared by dissolving a mixture of cationic lipid (e.g., CLinDMA or DOBMA, see structures and ratios for Formulations in Table 13), DSPC, Cholesterol, and PEG-DMG (ratios shown in Table 13) in absolute ethanol at a concentration of about 15 mg/mL. The nitrogen to phosphate ratio is approximate to 3: 1.
[0373] Equal volume of siNA/carrier and lipid solutions are delivered with two FPLC pumps at the same flow rates to a mixing T connector. A back pressure valve is used to adjust to the desired particle size. The resulting milky mixture is collected in a sterile glass bottle. This mixture is then diluted slowly with an equal volume of citrate buffer, and filtered through an ion- exchange membrane to remove any free siNA/carrier in the mixture. Ultra filtration against citrate buffer (pH 4.0) is employed to remove ethanol (test stick from ALCO screen), and against PBS (pH 7.4) to exchange buffer. The final LNP is obtained by concentrating to a desired volume and sterile filtered through a 0.2 μιη filter. The obtained LNPs are characterized in term of particle size, Zeta potential, alcohol content, total lipid content, nucleic acid encapsulated, and total nucleic acid concentration.
LNP Manufacture Process
[0374] In a non-limiting example, a LNP-086 siNA/carrier formulation is prepared in bulk as follows. The process consists of (1) preparing a lipid solution; (2) preparing an siNA/carrier solution; (3) mixing/particle formation; (4) incubation; (5) dilution; (6) ultrafiltration and concentration.
1. Preparation of Lipid Solution SIR-ONC-00001
[0375] A 3-necked 2L round bottom flask, a condenser, measuring cylinders, and two 10L conical glass vessels are depyrogenated. The lipids are warmed to room temperature. Into the3- necked round bottom flask is transferred 50.44g of CLinDMA with a pipette and 43.32g of DSPC, 5.32g of Cholesterol , 6.96g of PEG-DMG, and 2.64g of linoleyl alcohol are added. To the mixture is added 1L of ethanol. The round bottom flask is placed in a heating mantle that is connected to a J-CHEM process controller. The lipid suspension is stirred under Argon with a stir bar and a condenser on top. A thermocouple probe is put into the suspension through one neck of the round bottom flask with a sealed adapter. The suspension is heated at 30 °C until it became clear. The solution is allowed to cool to room temperature and transferred to a conical glass vessel and sealed with a cap.
2. Preparation of siNA/Carrier Solution
[0376] Into a sterile container, such as the Corning storage bottle, is weighed 3.6 g times the water correction factor (approximately 1.2) of siNA-1 powder. The siNA is transferred to a depyrogenated 5 L glass vessel. The weighing container is rinsed 3x with citrate buffer (25mM, pH 4.0, and lOOmM NaCl) and the rinses are placed into the 5 L vessel, QS with citrate buffer to 4 L. The concentration of the siNA solution is determined with a UV spectrometer using the following procedure. 20 μΐ^ is removed from the solution, diluted 50 times to 1000 μί, and the UV reading recorded at A260 nm after blanking with citrate buffer. This is repeated. If the readings for the two samples are consistent, an average is taken and the concentration is calculated based on the extinction coefficients of the siNAs. If the final concentration are out of the range of 0.90 + 0.01 mg/mL, the concentration is adjusted by adding more siNA/carrier powder, or adding more citrate buffer. This process is repeated for the second siNA, siNA-2.. Into a depyrogenated 10L glass vessel, 4 L of each 0.9 mg/mL siNA solution is transferred.
[0377] Alternatively, if the siNA/carrier solution comprised a single siNA duplex and or carrier instead of a cocktail of two or more siNA duplexes and/or carriers, then the siNA/carrier is dissolved in 25 mM citrate buffer (pH 4.0, 100 mM of NaCl) to give a final concentration of 0.9 mg/mL.
[0378] The lipid/ethanol solution is then sterile/filtered through a Pall Acropak 20 0.8/0.2 μιη sterile filter PN 12203 into a depyrogenated glass vessel using a Master Flex Peristaltic SIR-ONC-00001
Pump Model 7520-40 to provide a sterile starting material for the encapsulation process. The filtration process is run at an 80 mL scale with a membrane area of 20 cm . The flow rate is 280 mL/min. This process is scaleable by increasing the tubing diameter and the filtration area.
3. Particle formation - Mixing step
[0379] An AKTA P900 pump is turned on and sanitized by placing 1000 mL of 1 N NaOH into a 1 L glass vessel and 1000 mL of 70% ethanol into a 1 L glass vessel and attaching the pump with a pressure lid to each vessel. A 2000 mL glass vessel is placed below the pump outlet and the flow rate is set to 40 mL/min for a 40 minute time period with argon flushing the system at 10 psi. When the sanitation is complete, the gas is turned off and the pump is stored in the solutions until ready for use. Prior to use, the pump flow is verified by using 200 mL of ethanol and 200 mL of sterile citrate buffer.
[0380] To the AKTA pump is attached the sterile lipid/ethanol solution, the sterile siNA/carrier or siNA/carrier cocktail /citrate buffer solution and a depyrogenated receiving vessel (2x batch size) with lid. The gas is turned on and the pressure maintained between 5 to 10 psi during mixing.
4. Incubation
[0381] The solution is held after mixing for a 22 + 2 hour incubation. The incubation is done at room temperature (20 - 25°C) and the in-process solution was protected from light.
5. Dilution
[0382] The lipid siNA solution is diluted with an equal volume of Citrate buffer using a dual head peristaltic pump, Master Flex Peristaltic Pump, Model 7520-40 that is set up with equal lengths of tubing and a Tee connection and a flow rate of 360 mL/minute.
6. Ultrafiltration and Concentration
[0383] The ultrafiltration process is a timed process and the flow rates must be monitored carefully. This is a two step process; the first is a concentration step taking the diluted material from 32 liters to 3600 mLs and to a concentration of approximately 2 mg/mL. SIR-ONC-00001
[0384] In the first step, a Flexstand with a ultrafiltration membrane GE PN UFP-100-C-35A installed is attached to the quatroflow pump. 200 mL of WFI is added to the reservoir followed by 3 liters of 0.5 N sodium hydroxide which is then flushed through the retentate to waste. This process is repeated three times. Then 3 L WFI are flushed through the system twice followed by 3 L of citrate buffer. The pump is then drained.
[0385] The diluted LNP solution is placed into the reservoir to the 4 liter mark. The pump is turned on and the pump speed adjusted so the permeate flow rate is 300 mL/min. and the liquid level is constant at 4L in the reservoir. The pump is stopped when all the diluted LNP solution has been transferred to the reservoir. The diluted LNP solution is concentrated to 3600 mL in 240 minutes by adjusting the pump speed as necessary.
[0386] The second step is a diafiltration step exchanging the ethanol citrate buffer to phosphate buffered saline. The diafiltration step takes 3 hours and again the flow rates must be carefully monitored. During this step, the ethanol concentration is monitored by head space GC. After 3 hours (20 diafiltration volumes), a second concentration is undertaken to concentrate the solution to approximately 6 mg/mL or a volume of 1.2 liters. This material is collected into a depyrogenated glass vessel. The system is rinsed with 400 mL of PBS at high flow rate and the permeate line closed. This material is collected and added to the first collection. The expected concentration at this point is 4.5 mg/mL. The concentration and volume are determined.
[0387] The feed tubing of the peristaltic pump is placed into a container containing 72 L of PBS (0.05 μιη filtered) and the flow rate is adjusted initially to maintain a constant volume of 3600 mL in the reservoir and then increased to 400 mL/min. The LNP solution is diafiltered with PBS (20 volumes) for 180 minutes.
[0388] The LNP solution is concentrated to the 1.2 liter mark and collected into a depyrogenated 2 L graduated cylinder. 400 mL of PBS are added to the reservoir and the pump is recirculated for 2 minutes. The rinse is collected and added to the collected LNP solution in the graduated cylinder. SIR-ONC-00001
[0389] The obtained LNPs are characterized in terms of particle size, Zeta potential, alcohol content, total lipid content, nucleic acid encapsulated, and total nucleic acid concentration.
[0390] One skilled in the art would readily appreciate that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The methods and compositions described herein, as presently representative of preferred embodiments, are exemplary and are not intended as limitations on the scope of the invention. Changes therein and other uses will occur to those skilled in the art, which are encompassed within the spirit of the invention, are defined by the scope of the claims.
SI -ONC-00001 -US-PSP
Table 10: MET Accession Numbers
NM_000245 - SEQ ID NO: 1339
Homo sapiens met proto-oncogene (hepatocyte growth factor receptor)
(MET), transcript variant 2, mRNA. NM_000245.2 01:42741654
NM_001168629
Macaca mulatta met proto-oncogene (MET), mRNA.
NM_001168629.1 01:274323301
NM_008591
Mus musculus met proto-oncogene (MET), mRNA.
NM 008591.2 01:146198695
NM_001111071
Ovis aries growth factor receptor c-met (MET), mRNA
NM 001111071.1 01:162287028
SIR-ONC-00001 -US-PSP
Table 11
Non-limiting examples of Stabilization Chemistries for chemically modified siNA constructs
Figure imgf000151_0001
SIR-ONC-00001 -US-PSP
Figure imgf000152_0001
SI -ONC-00001 -US-PSP
Figure imgf000153_0001
CAP = any terminal cap, see for example Figure 5.
All Stab chemistries can be used in combination with each other for duplexes of the invention
(e.g., as combinations of sense and antisense strand chemistries), or alternately can be used in isolation, e.g., for single stranded nucleic acid molecules of the invention.
All Stab chemistries can comprise 3 '-overhang nucleotides having 2'-0-alkyl, 2'-deoxy-2'- fluoro, 2'-deoxy, LNA or other modified nucleotides or non-nucleotides.
All Stab chemistries typically comprise about 19-21 nucleotides, but can vary as described herein.
All Stab chemistries can also include a single ribonucleotide in the sense or passenger strand at the 11th base paired position of the double- stranded nucleic acid duplex as determined from the 5 '-end of the antisense or guide strand (see Figure 4C).
S = sense strand.
AS = antisense strand
*Stab 23 has a single ribonucleotide adjacent to 3 '-CAP.
*Stab 24 and Stab 28 have a single ribonucleotide at 5'-terminus.
*Stab 25, Stab 26, Stab 27, Stab 35, Stab 35G*, Stab 35N*, Stab 35rev*, Stab 36, Stab 50*,
Stab53*, Stab 53N*, and Stab 54 have three ribonucleotides at 5'-terminus.
*Stab 29, Stab 30, Stab 31, Stab 33, and Stab 34 any purine at first three nucleotide positions from 5 '-terminus are ribonucleotides.
p = phosphorothioate linkage
†Stab 35 has 2'-0-methyl U at 3'-overhangs and three ribonucleotides at 5'-terminus †Stab 36 has 2'-0-methyl overhangs that are complementary to the target sequence
(naturally occurring overhangs) and three ribonucleotides at 5 '-terminus
$ - Stab 04H, Stab 06C, Stabl07H, Stab07mU, Stab09H, Stabl6C, Stab 16H, Stabl8C, Stab 18H, Stab 52H, Stab 05C, Stab05N, StablOC, StablON, Stab35G*, Stab35N*, Stab35N*, Stab35rev*, Stab 50*, Stab 53*, Stab 53N*, Stab 54 have two 2'-0-methyl U 3'-overhangs. Stab35G*, Stab 35N*, Stab35rev*, Stab50*, Stab53*, and Stab53N* do not allow for a 2"-0- methyl modification at position 14 of the guide strand as determined from position the 5'- end. SIR-ONC-00001 -US-PSP
Table 12
A. 2.5 μιηοΐ Synthesis Cycle ABI 394 Instrument
Figure imgf000154_0001
0.2 μιηοΐ Synthesis Cycle ABI 394 Instrument
Figure imgf000154_0002
SI -ONC-00001 -US-PSP
C. 0.2 μηιοΐ Synthesis Cycle 96 well Instrument
Figure imgf000155_0001
• Wait time does not include contact time during delivery.
• Tandem synthesis utilizes double coupling of linker molecule
SI -ONC-00001 -US-PSP
Table 13
Lipid Nanoparticle (LNP) Formulations
Figure imgf000156_0001
SI -ONC-00001 -US-PSP
Figure imgf000157_0001
SI -ONC-00001 -US-PSP
Figure imgf000158_0001
N/P ratio = Nitrogen:Phosphorous ratio between cationic lipid and nucleic acid
The 2KPEG utilized is PEG2000, a polydispersion which can typically vary from -1500 to -3000 Da (i.e., where PEG(n) is about 33 to about 67, or on average -45). SI -ONC-00001 -US-PSP
Table 14
CLinDMA structure
Figure imgf000159_0001
pCLinDMA structure
Figure imgf000159_0002
eCLinDMA structure
Figure imgf000159_0003
DEGCLinDMA structure
Figure imgf000159_0004
PEG-n-DMG structure
Figure imgf000159_0005
n = about 33 to 67, average = 45 for 2KPEG/PEG2000 SIR-ONC-00001 -US-PSP
DMOBA structure
Figure imgf000160_0001
DMLBA structure
Figure imgf000160_0002
DSPC structure
Figure imgf000160_0003
Cholesterol structure
Figure imgf000160_0004
SI -ONC-00001 -US-PSP
2KPEG-Cholesterol structure
Figure imgf000161_0001
n = about 33 to 67, average = 45 for 2KPEG/PEG2000
2KPEG-DMG structure
Figure imgf000161_0002
n = about 33 to 67, average = 45 for 2KPEG/PEG2000
COIM STRUCTURE
Figure imgf000161_0003
SI -ONC-00001 -US-PSP
5-CLIMAND 2-CLIM STRUCTURE
Figure imgf000162_0001
Figure imgf000162_0002
DLinDMA structure
Figure imgf000162_0003

Claims

SI -ONC-00001 -US-PSP
What we claim is:
A double- stranded short interfering nucleic acid (siNA) molecule that inhibits the expression of MET, wherein
(a) the siNA comprises a sense strand and an antisense strand;
(b) each strand is independently 15 to 30 nucleotides in length; and
(c) the antisense strand comprises at least 15 nucleotides having sequence complementary to any of:
5'-GUGGCACUUUACUUACUUU-3' (SEQ ID NO: 1);
5'-UUUACUUACUUUAACUGGA-3' (SEQ ID NO: 43);
5'-CAAGCAUCUUCAGUUACCG-3' (SEQ ID NO: 51); or
5 ' - AG A AGG A AGUGUUU A AUAU-3 ' (SEQ ID NO: 53).
A double- stranded short interfering nucleic acid (siNA) molecule that inhibits the expression of MET, wherein
(a) the siNA comprises a sense strand and an antisense strand;
(b) each strand is independently 15 to 30 nucleotides in length; and
(c) the antisense strand comprises at least a 15 nucleotide sequence of:
5'-AAAGUAAGUAAAGUGCCAC-3' (SEQ ID NO: 1049);
5'-UCCAGUUAAAGUAAGUAAA-3' (SEQ ID NO: 1091);
5'-CGGUAACUGAAGAUGCUUG-3' (SEQ ID NO: 1099); or
5'-AUAUUAAACACUUCCUUCU-3' (SEQ ID NO: 1101); and wherein one of more of the nucleotides are optionally chemically modified.
The double-stranded short interfering nucleic acid (siNA) molecule of claim 2, wherein the sense strand comprises at least a 15 nucleotide sequence of:
5'-GUGGCACUUUACUUACUUU-3' (SEQ ID NO: 1);
5'-UUUACUUACUUUAACUGGA-3' (SEQ ID NO: 43);
5'-CAAGCAUCUUCAGUUACCG-3' (SEQ ID NO: 51); or SI -ONC-00001 -US-PSP
5 ' - AG A AGG A AGUGUUU A AUAU-3 ' (SEQ ID NO: 53); and wherein one of more of the nucleotides are optionally chemically modified.
4. A double- stranded short interfering nucleic acid (siNA) molecule that inhibits the expression of MET, wherein the siNA comprises any of:
5'-GUGGCACUUUACUUACUUU-3' (SEQ ID NO: 1) and 5'- AA AGUA AGUA A AGUGCC AC-3 ' (SEQ ID NO: 1049); or 5'-UUUACUUACUUUAACUGGA-3' (SEQ ID NO: 43) and 5'- UCCAGUUAAAGUAAGUAAA-3' (SEQ ID NO: 1091); or 5'-CAAGCAUCUUCAGUUACCG-3' (SEQ ID NO: 51) and 5'- CGGU A ACUG A AG AUGCUUG-3 ' (SEQ ID NO: 1099); or 5'- AG A AGG A AGUGUUU A AUAU-3' (SEQ ID NO: 53) and 5'- AUAUUAAACACUUCCUUCU-3' (SEQ ID NO: 1101).
5. The double- stranded short interfering nucleic acid (siNA) molecule according to claims 1, 2, 3, or 4, wherein at least one nucleotide is a chemically modified nucleotide.
6. The double- stranded short interfering nucleic acid (siNA) molecule according to claims 1, 2, 3, or 4, further comprising at least one non-nucleotide.
7. The double- stranded short interfering nucleic acid (siNA) molecule according to claims 1, 2, 3, or 4, wherein at least one nucleotide comprises a universal base.
8. The double- stranded short interfering nucleic acid (siNA) molecule according to claims 1, 2, 3, or 4, having at least one phosphorothioate internucleotide linkage.
9. The double- stranded short interfering nucleic acid (siNA) molecule according to claims 1, 2, 3, or 4, comprising a cap on the 3'-end, 5'-end or both 3' and 5' ends of at least one strand.
10. The double- stranded short interfering nucleic acid (siNA) molecule according to claims 1, 2, 3, or 4, comprising one or more 3'- overhang nucleotides on one or both strands. SI -ONC-00001 -US-PSP
11. The double-stranded short interfering nucleic acid (siNA) molecule of claim 10, wherein the 3 '-overhang nucleotides on at least one strand are 2-O-methyl nucleotides.
12. The double-stranded short interfering nucleic acid (siNA) molecule of claim 11, wherein the 2-O-methyl nucleotides are linked with a phosphorothioate internucleotide linkage.
13. The double-stranded short interfering nucleic acid (siNA) molecule of claim 5, wherein the chemically modified nucleotide is a 2'-deoxy-2'-fluoro nucleotide.
14. The double-stranded short interfering nucleic acid (siNA) molecule of claim 5, wherein the chemically modified nucleotide is a 2'-deoxy nucleotide.
15. The double-stranded short interfering nucleic acid (siNA) molecule of claim 5, wherein the chemically modified nucleotide is a 2'-0-alkyl nucleotide.
16. The double- stranded short interfering nucleic acid (siNA) molecule according to claims 1, 2, or 4, wherein five or more pyrimidine nucleotides in one or both strands are 2'-deoxy-2'-fluoro pyrimidine nucleotides.
17. The double- stranded short interfering nucleic acid (siNA) molecule according to claims 1, 2, or 4, wherein five or more pyrimidine nucleotides in one or both strands are 2' -O-methyl pyrimidine nucleotides.
18. The double- stranded short interfering nucleic acid (siNA) molecule according to claims 1, 2, or 4, wherein five or more purine nucleotides in one or both strands are 2'-deoxy-2'-fluoro purine nucleotides.
19. The double- stranded short interfering nucleic acid (siNA) molecule according to claims 1, 2, or 4, wherein five or more purine nucleotides in one or both strands are 2' -O-methyl purine nucleotides.
20. The double-stranded short interfering nucleic acid (siNA) molecule of claim 16, wherein five or more purine nucleotides in one or both strands are 2' -O-methyl purine nucleotides.
21. The double-stranded short interfering nucleic acid (siNA) molecule of claim 17, wherein five or more purine nucleotides in one or both strands are 2'-deoxy-2'-fluoro nucleotides. SI -ONC-00001 -US-PSP
22. A double-stranded short interfering nucleic acid (siNA) molecule, comprising formula (A) having a sense strand and an antisense strand:
B NX3 (N)x2 B -3'
B (N)xi NX4 [N]X5 -5'
(A) wherein, the upper strand is the sense strand and the lower strand is the antisense strand of the double- stranded nucleic acid molecule; wherein the antisense strand comprises at least a 15 nucleotide sequence of SEQ ID NO: 1049, SEQ ID NO: 1091, SEQ ID NO: 1099, or SEQ ID NO: 1101, and the sense strand comprises a sequence having complementarity to the antisense strand; each N is independently a nucleotide which is unmodified or chemically modified or a non-nucleotide; each B is a terminal cap that is present or absent;
(N) represents overhanging nucleotides, each of which is independently unmodified or chemically modified;
[N] represents nucleotides that are ribonucleotides ; XI and X2 are independently integers from 0 to 4; X3 is an integer from 15 to 30; X4 is an integer from 9 to 30; and
X5 is an integer from 0 to 6, provided that the sum of X4 and X5 is 15-30.
23. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein
(a) one or more pyrimidine nucleotides in Νχ4 positions are independently 2'- deoxy-2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof;
(b) one or more purine nucleotides in Νχ4 positions are independently 2'-deoxy- 2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof; SI -ONC-00001 -US-PSP
(c) one or more pyrimidine nucleotides in Νχ3 positions are independently 2'- deoxy-2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof; and
(d) one or more purine nucleotides in Νχ3 positions are independently 2'-deoxy- 2'-fluoro nucleotides, 2'-0-alkyl nucleotides, 2'-deoxy nucleotides, ribonucleotides, or any combination thereof.
24. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein X5 is 3.
25. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein X5 is 0.
26. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein XI is 2 and X2 is 2.
27. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein X5 is 0, XI is 2 and X2 is 2.
28. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein X5 is 3, XI is 2 and X2 is 2.
29. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein one or more nucleotides has an universal base.
30. The double-stranded short interfering nucleic acid (siNA) molecule according to claim
22, wherein the nucleotide at position 14 from the 5 '-end of the antisense strand is not a 2'-0-methyl nucleotide.
31. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein one or more overhang nucleotides is a 2'-0-methyl nucleotide.
32. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, comprising at least one phosphorothioate internucleotide linkage.
33. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein X5 = 0, 1, 2, or 3; each XI and X2 = 1 or 2; X3 = 18, 19, 20, 21, 22, or 23, and X4 = 17, 18, 19, 20, 21, 22, or 23. SI -ONC-00001 -US-PSP
34. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein X5 = 3; each XI and X2 = 2; X3 = 19, and X4 = 16.
35. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 22, wherein X5 = 0; each XI and X2 = 2; X3 = 19, and X4 = 19.
36. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 23, wherein
(a) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ4 positions are 2'-deoxy-2'- fluoro nucleotides;
(b) 1, 2, 3, 4, 5 or more purine nucleotides in Νχ4 positions are 2'-0-alkyl nucleotides;
(c) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ3 positions are 2'-deoxy-2'- fluoro nucleotides; and
(d) 1, 2, 3, 4, 5 or more purine nucleotides in Νχ3 positions are 2'-deoxy nucleotides.
37. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 23, wherein
(a) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ4 positions are 2'-0-alkyl nucleotides;
(b) 1, 2, 3, 4, 5 or more purine nucleotides in Νχ4 positions are ribonucleotides;
(c) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ3 positions are 2'-0-alkyl nucleotides; and
(d) 1, 2, 3, 4, 5 or more purine nucleotides in Νχ3 positions are ribonucleotides.
38. The double-stranded short interfering nucleic acid (siNA) molecule according to claim 23, wherein
(a) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ4 positions are 2'-deoxy-2'- fluoro nucleotides;
(b) 1, 2, 3, 4, 5 or more purine nucleotides in Νχ4 positions are 2'-0-alkyl nucleotides;
(c) 1, 2, 3, 4, 5 or more pyrimidine nucleotides in Νχ3 positions are 2'-0-alkyl nucleotides; and SI -ONC-00001 -US-PSP
(d) 1, 2, 3, 4, 5 or more purine nucleotides in Νχ3 positions are 2'-deoxy-2'-fluoro nucleotides.
39. The double- stranded short interfering nucleic acid (siNA) molecule according to any of claims 36, 37 or 38, comprising one or more phosphorothioate internucleotide linkages.
40. A composition comprising the double- stranded short interfering nucleic acid (siNA) of any of claims 1 or 22 in a pharmaceutically acceptable carrier or diluent.
41. A method of treating a human subject suffering from a condition which is mediated by the action, or by loss of action, of MET, which comprises administering to said subject an effective amount of the double- stranded short interfering nucleic acid (siNA) molecule of claim 2.
42. The method according to claim 41, wherein the condition is cancer.
PCT/US2011/031247 2010-04-12 2011-04-05 RNA INTERFERENCE MEDIATED INHIBITION OF MET GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA) WO2011130065A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32328410P 2010-04-12 2010-04-12
US61/323,284 2010-04-12

Publications (1)

Publication Number Publication Date
WO2011130065A1 true WO2011130065A1 (en) 2011-10-20

Family

ID=44798966

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/031247 WO2011130065A1 (en) 2010-04-12 2011-04-05 RNA INTERFERENCE MEDIATED INHIBITION OF MET GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)

Country Status (1)

Country Link
WO (1) WO2011130065A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2520651A2 (en) * 2009-12-31 2012-11-07 Samyang Biopharmaceuticals Corporation Sirna for inhibiting c-met expression and an anti-cancer composition comprising the same
US20150105452A1 (en) * 2012-03-28 2015-04-16 Somalogic, Inc. Aptamers to PDGF and VEGF and Their Use in Treating PDGF and VEGF Mediated Conditions
US9695424B2 (en) 2013-09-09 2017-07-04 Somalogic, Inc. PDGF and VEGF aptamers having improved stability and their use in treating PDGF and VEGF mediated diseases and disorders
US9938314B2 (en) 2013-11-21 2018-04-10 Somalogic, Inc. Cytidine-5-carboxamide modified nucleotide compositions and methods related thereto
US10221207B2 (en) 2010-04-12 2019-03-05 Somalogic, Inc. 5-position modified pyrimidines and their use
CN115094059A (en) * 2021-11-04 2022-09-23 山东大学齐鲁医院 Application of biomarker in preparation of reagent for assisting in diagnosis of cancer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090099121A1 (en) * 2002-02-20 2009-04-16 Mcswiggen James RNA INTERFERENCE MEDIATED INHIBITION OF MATRIX METALLOPROTEINASE 13 (MMP13) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2009143281A2 (en) * 2008-05-20 2009-11-26 Intradigm Corporation Compositions comprising c-met sirna and methods of use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090099121A1 (en) * 2002-02-20 2009-04-16 Mcswiggen James RNA INTERFERENCE MEDIATED INHIBITION OF MATRIX METALLOPROTEINASE 13 (MMP13) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2009143281A2 (en) * 2008-05-20 2009-11-26 Intradigm Corporation Compositions comprising c-met sirna and methods of use thereof

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2520651A2 (en) * 2009-12-31 2012-11-07 Samyang Biopharmaceuticals Corporation Sirna for inhibiting c-met expression and an anti-cancer composition comprising the same
EP2520651A4 (en) * 2009-12-31 2013-07-31 Samyang Biopharmaceuticals Sirna for inhibiting c-met expression and anti-cancer comprising the same
US10221207B2 (en) 2010-04-12 2019-03-05 Somalogic, Inc. 5-position modified pyrimidines and their use
US9410156B2 (en) * 2012-03-28 2016-08-09 Somalogic, Inc. Aptamers to PDGF and VEGF and their use in treating PDGF and VEGF mediated conditions
US9701967B2 (en) 2012-03-28 2017-07-11 Somalogic, Inc. Aptamers to PDGF and VEGF and their use in treating PDGF and VEGF mediated conditions
US10221421B2 (en) 2012-03-28 2019-03-05 Somalogic, Inc. Post-selec modification methods
US20150105452A1 (en) * 2012-03-28 2015-04-16 Somalogic, Inc. Aptamers to PDGF and VEGF and Their Use in Treating PDGF and VEGF Mediated Conditions
US11208663B2 (en) 2012-03-28 2021-12-28 Somalogic, Inc. Post-selex modification methods
US9695424B2 (en) 2013-09-09 2017-07-04 Somalogic, Inc. PDGF and VEGF aptamers having improved stability and their use in treating PDGF and VEGF mediated diseases and disorders
US9994857B2 (en) 2013-09-09 2018-06-12 Somalogic, Inc. PDGF and VEGF aptamers having improved stability and their use in treating PDGF and VEGF mediated diseases and disorders
US10544419B2 (en) 2013-09-09 2020-01-28 Somalogic Inc. PDGF and VEGF aptamers having improved stability and their use in treating PDGF and VEGF mediated diseases and disorders
US9938314B2 (en) 2013-11-21 2018-04-10 Somalogic, Inc. Cytidine-5-carboxamide modified nucleotide compositions and methods related thereto
US10239908B2 (en) 2013-11-21 2019-03-26 Somalogic, Inc. Cytidine-5-carboxamide modified nucleotide compositions and methods related thereto
CN115094059A (en) * 2021-11-04 2022-09-23 山东大学齐鲁医院 Application of biomarker in preparation of reagent for assisting in diagnosis of cancer

Similar Documents

Publication Publication Date Title
US20230030119A1 (en) Rna interference mediated inhibition of catenin (cadherin-associated protein), beta 1 (ctnnb1) gene expression using short interfering nucleic acid (sina)
US9879262B2 (en) RNA interference mediated inhibition of hepatitis B virus (HBV) gene expression using short interfering nucleic acid (siNA)
AU2012268619B2 (en) RNA interference mediated inhibition of isocitrate dehydrogenase (IDH1) gene expression
WO2011130065A1 (en) RNA INTERFERENCE MEDIATED INHIBITION OF MET GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2012021383A2 (en) RNA INTERFERENCE MEDIATED INHIBITION OF MITOGEN-ACTIVATED PROTEIN KINASE 1 (MAPK1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
AU2019264591A1 (en) RNA interference mediated inhibition of hepatitis B virus (HBV) gene expression using short interfering nucleic acid (siNA)

Legal Events

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

Ref document number: 11769314

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11769314

Country of ref document: EP

Kind code of ref document: A1