US20110313016A1 - Treatment of Intestinal Conditions - Google Patents

Treatment of Intestinal Conditions Download PDF

Info

Publication number
US20110313016A1
US20110313016A1 US11/908,696 US90869606A US2011313016A1 US 20110313016 A1 US20110313016 A1 US 20110313016A1 US 90869606 A US90869606 A US 90869606A US 2011313016 A1 US2011313016 A1 US 2011313016A1
Authority
US
United States
Prior art keywords
sina
sirna
seq
sina molecule
disorder
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/908,696
Inventor
Ana I. Jiménez
Irene Gascön
Maria Concepción Jiménez
José P. Román
Angela Sesto
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sylentis SA
Original Assignee
Sylentis SA
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 Sylentis SA filed Critical Sylentis SA
Assigned to SYLENTIS S.A.U. reassignment SYLENTIS S.A.U. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SESTO, ANGELA, JIMENEZ, ANA I., JIMENEZ, MARIA CONCEPCION, GASCON, IRENE, ROMAN, JOSE P.
Publication of US20110313016A1 publication Critical patent/US20110313016A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61K31/7115Nucleic acids or oligonucleotides having modified bases, i.e. other than adenine, guanine, cytosine, uracil or thymine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/10Laxatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/04Amoebicides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to methods and compositions for the treatment of intestinal pathologies by means of intrarectal administration of the RNAi technology.
  • the compositions of the invention comprise short interfering nucleic acid molecules (siNA) and related compounds including, but not limited to, small-interfering RNAs (siRNA).
  • compositions of the invention can be used for the treatment of intestinal pathologies including: hyperproliferative diseases, in particular, colorectal cancer; autoimmune and inflammatory bowel diseases (IBD), in particular Crohn's disease; colitis, in particular ulcerative colitis; irritable bowel syndrome; infectious diseases of the intestine, such as pseudomembranous colitis, amebiasis or intestinal tuberculosis; colonic polyps; diverticular disease; constipation; intestinal obstruction; malabsorption syndromes; rectal diseases and diarrhoea.
  • IBD autoimmune and inflammatory bowel diseases
  • colitis in particular ulcerative colitis
  • irritable bowel syndrome infectious diseases of the intestine, such as pseudomembranous colitis, amebiasis or intestinal tuberculosis; colonic polyps; diverticular disease; constipation; intestinal obstruction; malabsorption syndromes; rectal diseases and diarrhoea.
  • intestinal conditions caused by increased levels of interleukin-12 (IL-12), a cytokine involved in type 1 helper T (Th1) cells immune response are to be treated by this approach: for example, autoimmune diseases and IBD.
  • IL-12 interleukin-12
  • Th1 type 1 helper T
  • Compositions and methods comprising siRNA and related compounds targeting IL12-p40 subunit and/or IL12-p35 subunit are provided for the treatment of diseases associated with over-expression of IL-12, in particular Crohn's disease.
  • RNA interference refers to the process of sequence-specific post-transcriptional gene silencing mediated by double-stranded RNA (dsRNA).
  • dsRNA double-stranded RNA
  • RNAi The process of RNAi begins when the enzyme DICER encounters dsRNA and chops it into pieces called small-interfering RNAs or siRNA.
  • This protein belongs to the RNase III nuclease family. A complex of proteins gathers up these siRNAs and uses their code as a guide to search out and destroy any RNAs in the cell with a matching sequence, such as target mRNA (see Bosher & Labouesse, 2000; and Akashi et al., 2001).
  • RNAi for gene knockdown
  • mammalian cells have developed various protective mechanisms against viral infections that could impede the use of this approach. Indeed, the presence of extremely low levels of viral dsRNA triggers an interferon response, resulting in a global non-specific suppression of translation, which in turn triggers apoptosis (Williams, 1997, Gil & Esteban, 2000).
  • dsRNA was reported to specifically inhibit three genes in the mouse oocyte and early embryo. Translational arrest, and thus a PKR response, was not observed as the embryos continued to develop (Wianny & Zernicka-Goetz, 2000).
  • Research at Ribopharma AG demonstrated the functionality of RNAi in mammalian cells, using short (20-24 base pairs) dsRNAs to switch off genes in human cells without initiating the acute-phase response. Similar experiments carried out by other research groups confirmed these results (Elbashir et al., 2001; Caplen et al., 2001).
  • RNAs short hairpin RNAs
  • siRNAs small RNAs
  • stRNAs small temporally regulated RNAs
  • RNAi has rapidly become a well recognized tool for validating (identifying and assigning) gene functions.
  • RNAi employing short dsRNA oligonucleotides will yield an understanding of the function of genes being only partially sequenced.
  • RNAs microRNAs
  • antiagomirs a class of specially engineered compounds that can effectively silence the action of microRNAs (miRNAs), non-coding pieces of RNA that regulate gene expression.
  • Interleukin-12 (1L-12) is a heterodimeric 70 kDa glycoprotein (IL12-p70) consisting of a 40 kDa subunit (designated IL12-p40) and a 35 kDa subunit (designated IL12p35) linked by disulfide bonds that are essential for the biological activity of IL-12.
  • IL-12 is a key cytokine that regulates cell-mediated immune responses and type 1 helper T (Th1) cells inflammatory reaction (Gately et al., 1998; Trinchieri, 1998).
  • Th1 type 1 helper T
  • the ability of IL-12 to strongly promote the development of Th1 cells makes it an ideal target for the treatment of Th1 cell-mediated diseases, such as autoimmune diseases and inflammatory bowel disease (IBD).
  • Th1 cell-mediated diseases such as autoimmune diseases and inflammatory bowel disease (IBD).
  • IBD Crohn's disease
  • TNF- ⁇ tumour necrosis factor ⁇
  • Crohn's disease causes inflammation in the small intestine.
  • the inflammation can cause pain and can make the intestines empty frequently, resulting in diarrhoea.
  • the most common symptoms of Crohn's disease are abdominal pain and diarrhoea, although rectal bleeding, weight loss and fever may also occur. Bleeding may be serious and persistent, leading to anaemia. Children with Crohn's disease may suffer delayed development and stunted growth.
  • mesalamine a substance that helps control inflammation.
  • Sulfasalazine is the most commonly used of these drugs.
  • Patients who do not benefit from it or who cannot tolerate it may be put on other mesalamine-containing drugs, generally known as 5-ASA agents, such as Asacol, Dipentum or Pentasa.
  • 5-ASA agents such as Asacol, Dipentum or Pentasa.
  • Possible side effects of mesalamine preparations include nausea, vomiting, diarrhoea and headache. Some patients take corticosteroids to control inflammation.
  • These drugs are the most effective for active Crohn's disease, but they can cause serious side effects, including greater susceptibility to infection. Drugs that suppress the immune system are also used to treat Crohn's disease.
  • azathioprine 6-mercaptopurine and a related drug, azathioprine.
  • Immunosuppressive agents work by blocking the immune reaction that contributes to inflammation. These drugs may cause side effects like nausea, vomiting, and diarrhoea and may lower a person's resistance to infection. Surgery to remove part of the intestine can help Crohn's disease but cannot cure it. Due to the side effects and the lack of effectiveness of the current treatments for Crohn's disease, researchers continue to look for more effective treatments.
  • Inhibiting the action of IL-12 has been shown to suppress development and clinical progression of disease in a multitude of experimental models of autoimmunity and chronic inflammation (Caspi, 1998). These models include experimental autoimmune encephalomyelitis (EAE), experimental autoimmune uveitis (EAU), collagen-induced arthritis (CIA), autoimmune nephritis, insulin-dependent diabetes mellitus (IDDM) and different models for IBD (Vandenbroeck et al., 2004). In these models, the role of endogenous IL-12 has been addressed by using IL-12p40 knockout mice or by administering anti-IL-12 antibodies.
  • EAE experimental autoimmune encephalomyelitis
  • EAU experimental autoimmune uveitis
  • CIA collagen-induced arthritis
  • IDDM insulin-dependent diabetes mellitus
  • IBD insulin-dependent diabetes mellitus
  • mice with trinitrobenzene sulfonate-induced colitis have a Th1-mediated gut inflammation characterized by greatly increased production of IL-12, interferon- ⁇ and tumour necrosis factor ⁇ (TNF- ⁇ ).
  • administration of a monoclonal antibody against IL-12 can result in the resolution of established colitis and, if given at the time of induction of colitis, can prevent inflammation (Neurath et al., 1995).
  • Anti-interleukin-12 can also prevent and treat the spontaneous colitis seen in models of Th1-mediated inflammation such as mice that over-express the human CD3e gene and mice deficient in interleukin-10 (Davidson et al., 1998; Simpson et al., 1998).
  • siRNA targeting of IL-12 expression has already been used to obtain modified dendritic cells (DC) that might be used in a variety of therapeutic in vitro, ex vivo and in vivo methods to modulate T cell activity, and thus have use in therapeutic approaches for the treatment of immune disorders in a mammalian subject (WO 03/104455; Hill et al., 2003).
  • siRNA targeting of IL-12 expression in mature DC has revealed a critical role for IL-12 in natural killer cell interferon ⁇ (IFN- ⁇ ) secretion promoted by mature DC (Borg et al., 2004).
  • IFN- ⁇ natural killer cell interferon ⁇
  • IL-12 p35 inhibitors including siRNA have surprisingly demonstrated to block differentiation of preadipocytes to adipocytes and triglyceride accumulation in adipocytes (WO 03/104495).
  • siRNA targeting IL-12p40 has successfully been delivered by means of liposome encapsulation to murine peritoneal cavity to modulate the local and systemic inflammatory response after endotoxin challenge (Flynn et al., 2004).
  • siRNA targeting IL-12p40 has successfully been delivered by means of liposome encapsulation to murine peritoneal cavity to modulate the local and systemic inflammatory response after endotoxin challenge (Flynn et al., 2004).
  • intrarectal administration of siRNA for the downregulation of IL-12 nor of any other gene involved in intestinal pathologies.
  • compositions of the invention comprise short interfering nucleic acid molecules (siNA) and related compounds including, but not limited to, siRNA.
  • siNA short interfering nucleic acid molecules
  • compositions of the invention can be used in the preparation of a medicament for the treatment of intestinal pathologies including: hyperproliferative diseases, in particular, colorectal cancer; autoimmune and inflammatory bowel diseases (1BD), in particular Crohn's disease; colitis, in particular ulcerative colitis; irritable bowel syndrome; infectious diseases of the intestine, such as pseudomembranous colitis, amebiasis or intestinal tuberculosis; colonic polyps; diverticular disease; constipation; intestinal obstruction; malabsorption syndromes; rectal diseases and diarrhoea.
  • hyperproliferative diseases in particular, colorectal cancer
  • autoimmune and inflammatory bowel diseases (1BD) in particular Crohn's disease
  • colitis in particular ulcerative colitis
  • irritable bowel syndrome infectious diseases of the intestine, such as pseudomembranous colitis, amebiasis or intestinal tuberculosis; colonic polyps; diverticular disease; constipation; intestinal obstruction; malabsorption syndromes
  • siNA short interfering RNA
  • dsRNA double-stranded RNA
  • miRNA micro-RNA
  • antagomirs short hairpin RNA molecules capable of mediating RNA interference.
  • shRNA short hairpin RNA
  • the methods of the invention comprise the administration to a patient in the need thereof of an effective amount of one or more siNA of the invention for the treatment of an intestinal condition.
  • the methods of the invention comprise intrarectal administration of the therapeutic siNA.
  • the present invention relates to siNA or similar chemically synthesized entities that are directed at interfering with the mRNA expression of either the p35 or the p40 subunits of the cytokine IL-12, and that ultimately modulate the amount of protein produced.
  • Compositions and methods comprising above-mentioned siRNA and related compounds are intended for the treatment of diseases associated with over-expression of IL-12, such as autoimmune diseases and inflammatory bowel diseases (IBD), in particular, Crohn's disease.
  • diseases associated with over-expression of IL-12 such as autoimmune diseases and inflammatory bowel diseases (IBD), in particular, Crohn's disease.
  • IBD inflammatory bowel diseases
  • FIG. 1 Oligonucleotide sequences for siRNA molecules targeting IL-12 p35 and p40 subunits encompassed by the present invention.
  • the SEQ ID Nos given in the Figure refer to the sense (5′->3′) strand; typically sIRNA will be administered as dsRNA, so will include both the sense strand and its complement.
  • FIG. 2 Effect of siRNA on IL-12 p35 subunit expression in an in vitro system.
  • siRNA treatment reduces the levels of IL-12 p35 gene transcript.
  • RNA was prepared from SW480 cells treated with siRNAs for different times. The samples were analyzed by RT-PCR using specific primers. The values show the mean expression levels of different transcripts normalized to 18S as housekeeping gene.
  • FIG. 3 Effect of siRNA on IL-12 p40 subunit expression in an in vitro system.
  • A siRNA treatment reduces the levels of IL-12 p40 gene transcript in human cells.
  • RNA was prepared from SW480 cells treated with siRNA SEQ ID 67 and SEQ ID 79 at different times, at dose treatment of 200 nM. The values show the mean expression levels of different transcripts normalized to 18S as housekeeping gene. The values represent the mean of the percentage of the normalized mRNA levels upon siRNA interference over the control gene expression and their medium standard deviations (SEM).
  • B siRNA treatment reduces the levels of IL-12 p40 gene transcript in murine cells.
  • RNA was prepared from C2C12 cells treated with siRNA SEQ ID 86 and SEQ ID 87 at different times, at dose treatment of 100 nM.
  • SEQ ID 86 which is homologous to human SEQ ID 67, targets the mouse IL-12 p40 subunit. Further targeting the mouse IL-12 p40 subunit, SEQ ID 87 is the siRNA with the best score in mouse, and has no homologous siRNA duplex in human.
  • siNA molecules SEQ ID 86 and SEQ ID 87 are as described below, with 2 thymidine nucleotide 3′ overhangs. The values represent the mean of the percentage of the normalized mRNA levels compared to 18S upon siRNA interference over the control gene expression and their medium standard deviations (SEM).
  • FIG. 4 siRNA treatment reduces the levels of GFP gene transcript in small intestine.
  • the collected tissue in OCT was analyzed by microscopy and measured by photoshop program.
  • Data show single dose siRNA treatment (mice 2-3) and repeated dose treatment (mice 4-5).
  • the values show the expression levels of 25 representative images per mouse referred to control untreated mouse. Standard deviation of the data is represented.
  • FIG. 5 siRNA treatment reduces the levels of GFP gene transcript in small intestine.
  • the tissue collected in RNA later was analyzed by RT-PCR. Data show single dose siRNA treatment (mice 2-3) and repeated dose treatment (mice 4-5). Standard deviation is represented.
  • FIG. 6 siRNA treatment reduces the levels of GFP gene transcript in large intestine.
  • the tissue collected in OCT was analyzed by microscopy and measured by photoshop program.
  • Data show single dose siRNA treatment (mice 2-3) and repeated dose treatment (mice 4-5).
  • the values show the expression levels of 25 representative images per mouse referred to control untreated mouse. Standard deviation of the data is represented.
  • FIG. 7 siRNA treatment reduces the levels of GFP gene transcript in large intestine.
  • FIG. 8 Data of samples collected in OCT medium.
  • FIG. 9 Data of samples collected in RNA later.
  • the present invention relates to methods and compositions for the treatment of intestinal pathologies by means of intrarectal administration of the RNAi technology.
  • the compositions of the invention comprise short interfering nucleic acid molecules (siNA) that modulate the expression of target genes associated with conditions of the intestinal wall.
  • siNA short interfering nucleic acid molecules
  • the methods of the invention comprise the administration to a patient in need thereof of an effective amount of one or more siNA of the invention.
  • a gene is “targeted” by siNA according to the invention when, for example, the siNA selectively decrease or inhibit the expression of the gene.
  • siNA target a gene when the siNA hybridize under stringent conditions to the gene transcript.
  • siNA can be tested either in vitro or in vivo for the ability to target a gene.
  • a short fragment of the target gene sequence (e.g., 19-40 nucleotides in length) is chosen as the sequence of the siNA of the invention.
  • the siNA is siRNA.
  • the short fragment of target gene sequence is a fragment of the target gene mRNA.
  • the criteria for choosing a sequence fragment from the target gene mRNA to be a candidate siRNA molecule include: 1) a sequence from the target gene mRNA that is at least 50-100 nucleotides from the 5′ or 3′ end of the native mRNA molecule; 2) a sequence from the target gene mRNA that has a G/C content of between 30% and 70%, most preferably around 50%; 3) a sequence from the target gene mRNA that does not contain repetitive sequences (e.g., AAA, CCC, GGG, TTT, AAAA, CCCC, GGGG, TTTT); 4) a sequence from the target gene mRNA that is accessible in the mRNA; and 5) a sequence from the target gene mRNA that is unique to the target gene.
  • the sequence fragment from the target gene mRNA may meet one or more of the above-mentioned identified criteria.
  • the siRNA has a G/C content below 60% and/or lacks repetitive sequences.
  • the gene of interest is introduced as a nucleotide sequence in a prediction program that takes into account all the variables described above for the design of optimal oligonucleotides.
  • This program scans any mRNA nucleotide sequence for regions susceptible to be targeted by siRNA.
  • the output of this analysis is a score of possible siRNA oligonucleotides.
  • the highest scores are used to design double stranded RNA oligonucleotides (typically 21 bp long, although other lengths are also possible) that are typically made by chemical synthesis.
  • Candidate oligonucleotides can further be filtered for interspecies sequence conservation in order to facilitate the transition from animal to human clinical studies.
  • siNA molecules may be used to target homologous regions.
  • WO2005/045037 describes the design of siNA molecules to target such homologous sequences, for example by incorporating non-canonical base pairs, for example mismatches and/or wobble base pairs, that can provide additional target sequences.
  • non-canonical base pairs for example, mismatches and/or wobble bases
  • non-canonical base pairs can be used to generate siNA molecules that target more than one gene sequence.
  • non-canonical base pairs such as UU and CC base pairs are used to generate siNA molecules that are capable of targeting sequences for differing targets that share sequence homology.
  • siNAs of the invention are designed to include nucleic acid sequence that is complementary to the nucleotide sequence that is conserved between homologous genes.
  • a single siNA can be used to inhibit expression of more than one gene instead of using more than one siNA molecule to target different genes.
  • Sequence identity may be calculated by sequence comparison and alignment algorithms known in the art (see Gribskov and Devereux, Sequence Analysis Primer, Stockton Press, 1991, and references cited therein) and calculating the percent difference between the nucleotide sequences by, for example, the Smith-Waterman algorithm as implemented in the BESTFIT software program using default parameters (e.g., University of Wisconsin Genetic Computing Group). Greater than 90%, 95%, or 99% sequence identity between the siNA and the portion of the target gene is preferred.
  • the complementarity between the siNA and native RNA molecule may be defined functionally by hybridisation as well as functionally by its ability to decrease or inhibit the expression of a target gene. The ability of a siNA to affect gene expression can be determined empirically either in vivo or in vitro.
  • Preferred siNA molecules of the invention are double stranded.
  • double stranded siNA molecules comprise blunt ends.
  • double stranded siNA molecules comprise overhanging nucleotides (e.g., 1-5 nucleotide overhangs, preferably 2 nucleotide overhangs).
  • the overhanging nucleotides are 3′ overhangs.
  • the overhanging nucleotides are 5′ overhangs. Any type of nucleotide can be a part of the overhang.
  • the overhanging nucleotide or nucleotides are ribonucleic acids.
  • the overhanging nucleotide or nucleotides are deoxyribonucleic acids. In a preferred embodiment, the overhanging nucleotide or nucleotides are thymidine nucleotides. In another embodiment, the overhanging nucleotide or nucleotides are modified or non-classical nucleotides. The overhanging nucleotide or nucleotides may have non-classical internucleotide bonds (e.g., other than phosphodiester bond).
  • siNA can be synthesized by any method known in the art.
  • RNAs are preferably chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer.
  • siRNA can be obtained from commercial RNA oligo synthesis suppliers, including, but not limited to, Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, Colo., USA), Glen Research (Sterling, Va., USA), ChemGenes (Ashland, Mass., USA), and Cruachem (Glasgow, UK), Qiagen (Germany), Ambion (USA) and Invitrogen (Scotland).
  • siNA molecules of the invention can be expressed in cells by transfecting the cells with vectors containing the reverse complement siNA sequence under the control of a promoter. Once expressed, the siNA can be isolated from the cell using techniques well known in the art.
  • RNA oligo 50 ⁇ M solution 30 ⁇ l of each RNA oligo 50 ⁇ M solution are to be combined in 100 mM potassium acetate, 30 mM HEPES-KOH pH 7.4, 2 mM magnesium acetate. The solution is then incubated for 1 minute at 90° C., centrifuged for 15 seconds, and incubated for 1 hour at 37° C.
  • siRNA is a short hairpin RNA (shRNA); the two strands of the siRNA molecule may be connected by a linker region (e.g., a nucleotide linker or a non-nucleotide linker).
  • a linker region e.g., a nucleotide linker or a non-nucleotide linker.
  • the siNAs of the invention may contain one or more modified nucleotides and/or non-phosphodiester linkages. Chemical modifications well known in the art are capable of increasing stability, availability, and/or cell uptake of the siNA. The skilled person will be aware of other types of chemical modification which may be incorporated into RNA molecules (see International Publications WO03/070744 and WO2005/045037 for an overview of types of modifications).
  • modifications can be used to provide improved resistance to degradation or improved uptake.
  • modifications include phosphorothioate internucleotide linkages, 2′-O-methyl ribonucleotides (especially on the sense strand of a double stranded siRNA), 2′-deoxy-fluoro ribonucleotides, 2′-deoxy ribonucleotides, “universal base” nucleotides, 5-C-methyl nucleotides, and inverted deoxyabasic residue incorporation (see generally GB2406568).
  • modifications can be used to enhance the stability of the siRNA or to increase targeting efficiency.
  • Modifications include chemical cross linking between the two complementary strands of an siRNA, chemical modification of a 3′ or 5′ terminus of a strand of an siRNA, sugar modifications, nucleobase modifications and/or backbone modifications, 2′-fluoro modified ribonucleotides and 2′-deoxy ribonucleotides (see generally International Publication WO2004/029212).
  • modifications can be used to increase or decrease affinity for the complementary nucleotides in the target mRNA and/or in the complementary siNA strand (see generally International Publication WO2005/044976).
  • an unmodified pyrimidine nucleotide can be substituted for a 2-thio, 5-alkynyl, 5-methyl, or 5-propynyl pyrimidine.
  • an unmodified purine can be substituted with a 7-deza, 7-alkyl, or 7-alkenyl purine.
  • the siNA when the siNA is a double-stranded siRNA, the 3′-terminal nucleotide overhanging nucleotides are replaced by deoxyribonucleotides (see generally Elbashir et al., 2001).
  • the cells used for the experiments were human SW480 cells and murine muscle cells C2C12. After incubation of the cells with the corresponding siRNA duplexes, the levels of p35 and p40 expression were analyzed.
  • siRNA knockdown to specific phenotypes in cultured cells, it is necessary to demonstrate the decrease of the targeted protein or at least to demonstrate the reduction of the targeted mRNA.
  • mRNA levels of the target gene can be quantitated by real time PCR(RT-PCR). Further, the protein levels can be determined in a variety of ways well known in the art, such as Western blot analysis with specific antibodies to the different target allow direct monitoring of the reduction of targeted protein.
  • siRNA are introduced into cells by means of any transfection technique well known in the art.
  • a single transfection of siRNA duplex can be performed, for instance, by using a cationic lipid, such as Lipofectamine 2000 Reagent (Invitrogen), followed by an assay of silencing efficiency 24, 48 and 72 hours after transfection.
  • a cationic lipid such as Lipofectamine 2000 Reagent (Invitrogen)
  • a typical transfection protocol can be performed as follows: for one well of a E-well plate, we transfect using 100 nM for murine C2C12 cells or 200 nM for human SW480 cells as final concentration of siRNA. Following Lipofectamine 2000 Reagent protocol, the day before transfection, we seed 2-4 ⁇ 10 5 cells per well in 3 ml of an appropriate growth medium, containing DMEM, 10% serum, antibiotics and glutamine, and incubate cells under normal growth conditions (37° C. and 5% CO 2 ). On the day of transfection, cells have to be at 30-50% confluence.
  • the efficiency of transfection may depend on the cell type, but also on the passage number and the confluency of the cells.
  • the time and the manner of formation of siRNA-liposome complexes are also critical. Low transfection efficiencies are the most frequent cause of unsuccessful silencing. Good transfection is a non-trivial issue and needs to be carefully examined for each new cell line to be used.
  • Transfection efficiency may be tested transfecting reporter genes, for example a CMV-driven EGFP-expression plasmid (e.g. from Clontech) or a B-Gal expression plasmid, and then assessed by phase contrast and/or fluorescence microscopy the next day.
  • a knock-down phenotype may become apparent after 1 to 3 days, or even later.
  • depletion of the protein may be observed by immunofluorescence or Western blotting.
  • RNA fractions extracted from cells are pre-treated with DNase I and used for reverse transcription using a random primer.
  • PCR-amplified with a specific primer pair covering at least one exon-exon junction is used as control for amplification of pre-mRNAs.
  • RT-PCR of a non-targeted mRNA is also needed as control. Effective depletion of the mRNA yet undetectable reduction of target protein may indicate that a large reservoir of stable protein may exist in the cell.
  • RT-PCR amplification can be used to test in a more precise way the mRNA decrease or disappearance.
  • RT-PCR quantitates the initial amount of the template most specifically, sensitively and reproducibly.
  • RT-PCR monitors the fluorescence emitted during the reaction as an indicator of amplicon production during each PCR cycle, in a light cycler apparatus. This signal increases in direct proportion to the amount of PCR product in a reaction. By recording the amount of fluorescence emission at each cycle, it is possible to monitor the PCR reaction during exponential phase where the first significant increase in the amount of PCR product correlates to the initial amount of target template.
  • the PCR conditions were an initial step of 30 s at 95° C., followed by 40 cycles of 5 s at 95° C., 10 s at 62° C. and 15 s at 72° C.
  • Quantification of 18S mRNA was used as a housekeeping gene as a control for data normalization. Relative gene expression comparisons work best when the gene expression of the chosen endogenous/internal control is more abundant and remains constant, in proportion to total RNA, among the samples.
  • RNA target By using an invariant endogenous control as an active reference, quantitation of an mRNA target can be normalised for differences in the amount of total RNA added to each reaction.
  • the amplification curves obtained with the light cycler were analyzed in combination with the control kit DNA, which targets in vitro transcribed beta-globin DNA template, according to the manufacturer protocol.
  • a melting curve analysis was performed. The resulting melting curves allow discrimination between primer-dimers and specific PCR product.
  • GFP C57BL/6-TG mice This transgenic mouse line was bought from “The Jackson Laboratory”. Transgenic mice have been used because homozygous mice for this transgene die within the first two weeks following birth.
  • the transgenic mouse line with an “enhanced” GFP (EGFP) cDNA under the control of a chicken beta-actin promoter and cytomegalovirus enhancer makes all of the tissues, with the exception of erythrocytes and hair, appear green under excitation light.
  • This strain was generated in C57BL/6 mice.
  • the strain cDNA encoding enhanced green fluorescent protein (EGFP) was adjoined to the chicken beta actin promoter and cytomegalovirus enhancer.
  • a bovine globin polyadenylation signal was also included in the construct.
  • the EcoR1 sites included in the PCR primers were used to introduce the amplified EGFP cDNA into a pCAGGS expression vector containing the chicken beta-actin promoter and cytomegalovirus enhancer, beta-actin intron and bovine globin poly-adenylation signal.
  • the entire insert with the promoter and coding sequence was excised with Bam-HI and SalI and gel-purified.
  • siRNA duplex used for intrarectal injection in mice was purchased from Dharmacon.
  • Dharmacon Research Inc (Lafayette, Colo.) have developed a new generation of modified siRNA for in vivo use as a therapeutic, named siSTABLEv2.
  • Dharmacon's siSTABLEv2 siRNA have demonstrated an enhanced stability in serum with respect to that of non-modified siRNA.
  • Conventional siRNA are typically degraded within minutes in serum-containing environments, making in vivo use of siRNA problematic.
  • the siSTABLEv2 modification dramatically extends the siRNA stability in serum as described in Dharmacon's web page (http://www.dharmacon.com/docs/siSTABLE %20v2%20Flier.pdf).
  • the siRNA used to downregulate EGFP mRNA expression targeted the following sequence in EGFP mRNA: 5′-GGC UAC GUC CAG GAG CGC ACC-3′ (SEQ ID No 88).
  • the sense strand of the siRNA duplex was 5′-P GGC UAC GUC CAG CGC ACC-3′ (SEQ ID No 89) and the antisense strand was 5′-P U GCG CUC CUG GAC GUA GCC UU-3′ (SEQ ID No 90). This sequence is distributed by Dharmacon as pre-synthesized control siRNA green fluorescent protein duplex.
  • mice Males, 8 weeks old mice were used. The animals were kept in cages with free access to food and water until one day before the experimental protocol.
  • intrarectal therapeutic silencing mice were fasted for one day prior to the treatment.
  • the drugs are typically administered by injecting a small volume (120 ⁇ L) in the rectum.
  • Control mouse is treated with the vehicle alone.
  • animals were sacrificed two days after the first injection by cervical dislocation.
  • the protocol for the siRNA application in mouse is as follows. For each experimental administration, 60 ⁇ l siRNA duplexes were premixed with 60 ⁇ l of NaCl (1.8% w/v) up to physiological levels. In all cases animals were sacrificed two days after the first injection.
  • mice 2 and 3 were treated intrarectally with one dose of 250 ⁇ g (19 nanomols) of the siRNA vs GFP, while mice 4 and 5 were treated with two doses of 125 ⁇ g of siRNA during two consecutive days.
  • OCT blocks were storaged at ⁇ 80° C. until data processing.
  • OCT blocks were cut in slices of 12 ⁇ m by a cryostat (Leica CM 1850) at ⁇ 20° C.
  • the collected slices were analyzed on a fluorescence microscope (Olympus BX51) coupled to a digital camera (DP70), using a filter of 488 nm.
  • the sensitive conditions (ISO200), resolution image size (2040 ⁇ 1536) and time exposure (1 second) were set up for all the samples in order to be compared between them.
  • Green fluorescence was measured as an index of GFP expression by an Adobe Photoshop program (version 8.0).
  • the present invention may comprise the administration of one or more species of siNA molecule simultaneously. These species may be selected to target one or more target genes.
  • a single type of siNA is administered in the therapeutic methods of the invention.
  • a siNA of the invention is administered in combination with another siNA of the invention and/or with one or more other non-siNA therapeutic agents useful in the treatment, prevention or management of a disease condition of the intestine wall.
  • the term “in combination with” is not limited to the administration of therapeutic agents at exactly the same time, but rather it is meant that the siNAs of the invention and the other agent are administered to a patient in a sequence and within a time interval such that the benefit of the combination is greater than the benefit if they were administered otherwise.
  • each therapeutic agent may be administered at the same time or sequentially in any order at different points in time; however, if not administered at the same time, they should be administered sufficiently close in time so as to provide the desired therapeutic effect.
  • Each therapeutic agent can be administered separately, in any appropriate form and by any suitable route.
  • siNAs of the invention may be formulated into pharmaceutical compositions by any of the conventional techniques known in the art (see for example, Alfonso, G. et al., 1995, in: The Science and Practice of Pharmacy, Mack Publishing, Easton Pa., 19th ed.).
  • Formulations comprising one or more siNAs for use in the methods of the invention may be in numerous forms, and may depend on the various factors specific for each patient (e.g., the type and severity of disorder, type of siNA administered, age, body weight, response, and the past medical history of the patient), the number and type of siNAs in the formulation, the form of the composition (e.g., in liquid, semi-liquid or solid form), the therapeutic regime (e.g.
  • the therapeutic agent is administered over time as a slow infusion, a single bolus, once daily, several times a day or once every few days), and/or the route of administration (e.g., topical, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, or sublingual means).
  • route of administration e.g., topical, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, or sublingual means.
  • siNA molecules of the invention and formulations or compositions thereof may be administered directly or topically as is generally known in the art.
  • a siNA molecule can comprise a delivery vehicle, including liposomes, for administration to a subject.
  • Carriers and diluents and their salts can be present in pharmaceutically acceptable formulations.
  • 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 poly (lactic-co-glycolic) acid (PLGA) and PLCA microspheres, biodegradable nanocapsules, and bioadhesive microspheres, or by proteinaceous vectors.
  • nucleic acid 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
  • a siNA molecule of the invention may be complexed with membrane disruptive agents and/or a cationic lipid or helper lipid molecule.
  • Delivery systems which may be used with the invention include, for example, aqueous and non aqueous gels, creams, multiple emulsions, microemulsions, liposomes, ointments, aqueous and non aqueous solutions, lotions, aerosols, hydrocarbon bases and powders, and can contain excipients such as solubilizers, permeation enhancers (e.g., fatty acids, fatty acid esters, fatty alcohols and amino acids), and hydrophilic polymers (e.g., polycarbophil and polyvinylpyrolidone).
  • the pharmaceutically acceptable carrier is a liposome or a transdermal enhancer.
  • a pharmaceutical formulation of the invention is 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, or by injection. 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.
  • compositions prepared for storage or administration that include a pharmaceutically effective amount of the desired compounds in a pharmaceutically acceptable carrier or diluent.
  • Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art.
  • preservatives, stabilizers, dyes and flavouring agents can be provided. These include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid.
  • antioxidants and suspending agents can be used.
  • a pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, preferably all of the symptoms) of a disease state.
  • the pharmaceutically effective dose depends on the type of disease, the composition used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors that those skilled in the medical arts will recognize.
  • compositions of the invention can be administered in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and/or vehicles.
  • Formulations can be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • 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 monostearate 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 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 colouring agents, one or more flavouring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • colouring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavouring agents such as sucrose or saccharin.
  • 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 anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerin, glycerin, glycerin, glycerin, glycerin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol
  • 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 flavouring 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.
  • a sterile injectable preparation can also 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 anaesthetics, preservatives and buffering agents can be dissolved in the vehicle.
  • the composition can also be added to the animal feed or drinking water. It can be convenient to formulate the animal feed and drinking water compositions so that the animal takes in a therapeutically appropriate quantity of the composition along with its diet. It can also be convenient to present the composition as a premix for addition to the feed or drinking water.
  • nucleic acid molecules of the present invention can also be administered to a subject in combination with other therapeutic compounds to increase the overall therapeutic effect.
  • the use of multiple compounds to treat an indication can increase the beneficial effects while reducing the presence of side effects.
  • siNA molecules of the invention can be expressed within cells from eukaryotic promoters.
  • Recombinant vectors capable of expressing the siNA molecules can be delivered and persist in target cells.
  • vectors can be used that provide for transient expression of nucleic acid molecules. Such vectors can be repeatedly administered as necessary.
  • the siNA molecule interacts with the target mRNA and generates an RNAi response. Delivery of siNA molecule expressing vectors can be systemic, such as by intravenous or intramuscular 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.
  • GenBank Accession numbers corresponding to IL-12 p35 (Interleukin 12A, natural killer cell stimulatory factor 1, cytotoxic lymphocyte maturation factor 1, p35) and p40 (Interleukin 12B, natural killer cell stimulatory factor 2, cytotoxic lymphocyte maturation factor 2, p40) subunits are NM 000882 and NM 002187, respectively.
  • siNA molecules directed to target IL-12 p35 and p40 subunits were obtained.
  • the output of this analysis was a score of possible siNA oligonucleotides, the highest scores being used to design double stranded RNA oligonucleotides (typically 19 bp long) that were typically made by chemical synthesis.
  • siNA compositions of the invention are any of SEQ ID NOS:1-81 of FIG. 1 ; typically administered as a duplex of the sense strand and the antisense strand.
  • the invention also encompasses siNA that are 40 nucleotides or less and comprise a nucleotide sequence of any of SEQ ID NOS:1-81.
  • the siNA is 21-30 nucleotides long and comprises any one of SEQ ID NOS:1-81 of FIG. 1 . All siNA molecules used in the experiments described below were designed to have a 2 thymidine nucleotide 3′ overhang.
  • siRNA contained within FIG. 1 has been analysed in cell cultures. SiRNA with the best characteristics were selected to be tested and were applied to proper cell cultures, such as SW480. The effect of siRNA over the target gene was analyzed by RT-PCR according to the manufacturer's protocol. The gene target transcript levels were normalized using 18S as housekeeping gene. Some of the different siRNA that were tested and their different efficacies in the interference of the target gene are included in the FIG. 2 . These results correspond to SEQ ID 8 and SEQ ID 17 of FIG. 1 in SW480 cells expressing p35.
  • the values represent the mean of the percentage of the normalized mRNA levels upon siRNA interference over the control gene expression and their medium standard deviations (SEM).
  • SEM medium standard deviations
  • siRNA contained within FIG. 1 has been analyzed.
  • the siRNA with the best characteristics designed as described before, were tested in human and murine cells.
  • the p40 transcript level was analyzed by RT-PCR and normalized using 18S as housekeeping gene. These results correspond to SEQ ID 67, SEQ ID 79 in SW480 cells expressing p40 ( FIG. 3A ); and SEQ ID 86 and SEQ ID 87 in C2C12 cells expressing p40 ( FIG. 3B ).
  • siNA molecules SEQ ID 86 and SEQ ID 87 are as described in the figure, with 2 thymidine nucleotide 3′ overhangs.
  • siRNA corresponding to SEQ ID 67 in SW480, up to 65% compared to control cells.
  • siRNA corresponding to SEQ ID 86 decreased the gene expression to 61% at 48 h compared to the control. It is important to note that SEQ ID 67 and SEQ ID 86 correspond to homologous regions of human and mouse IL-12 p40 gene, respectively.
  • the siRNA application is made in order to determine the proper siRNA delivery in the intestine.
  • small intestine samples were collected in OCT medium and analyzed as previously described. Since the goal is to determine the downregulation of GFP gene transcript, levels of fluorescence were measured following siRNA application. No secondary effects were observed in the animals during the experimental protocols.
  • the first group of work (animals 2 and 3) was treated with a single dose of 250 ⁇ g of siRNA and sacrificed 48 h later.
  • the results indicate a significant decrease of fluorescence when compared with the control mouse.
  • the siRNA 250 ⁇ g
  • the siRNA is administered in two doses of 125 ⁇ g and analyzed 48 h after the first injection
  • the decrease of GFP expression was similar to that after a single application.
  • the results are shown in FIG. 4 . For each experimental condition an average of the data is represented.
  • the administered dose of 250 ⁇ g of siRNA in one or two applications was enough and sufficient to downregulate the level of GFP mRNA in small intestine, confirming the delivery of the siRNA in small intestine by intrarectal administration.
  • the level of downregulation compared to the control is higher when the analysis is done by RT-PCR, this being due to the higher sensitivity of the technique.
  • FIGS. 8 and 9 Data of samples collected in OCT medium and in RNA later are summarized in FIGS. 8 and 9 respectively.

Abstract

Methods and compositions for the treatment of intestinal disorders, such as IBD and Crohn's disease, are disclosed. Preferred compositions include siNA. Also disclosed is a method of specifically targeting siNA to treat intestinal disorders by intrarectal administration of siNA compounds.

Description

    FIELD OF THE INVENTION
  • The present invention relates to methods and compositions for the treatment of intestinal pathologies by means of intrarectal administration of the RNAi technology. The compositions of the invention comprise short interfering nucleic acid molecules (siNA) and related compounds including, but not limited to, small-interfering RNAs (siRNA). In particular, the compositions of the invention can be used for the treatment of intestinal pathologies including: hyperproliferative diseases, in particular, colorectal cancer; autoimmune and inflammatory bowel diseases (IBD), in particular Crohn's disease; colitis, in particular ulcerative colitis; irritable bowel syndrome; infectious diseases of the intestine, such as pseudomembranous colitis, amebiasis or intestinal tuberculosis; colonic polyps; diverticular disease; constipation; intestinal obstruction; malabsorption syndromes; rectal diseases and diarrhoea.
  • In certain embodiments, intestinal conditions caused by increased levels of interleukin-12 (IL-12), a cytokine involved in type 1 helper T (Th1) cells immune response are to be treated by this approach: for example, autoimmune diseases and IBD. Compositions and methods comprising siRNA and related compounds targeting IL12-p40 subunit and/or IL12-p35 subunit are provided for the treatment of diseases associated with over-expression of IL-12, in particular Crohn's disease.
  • BACKGROUND OF THE INVENTION RNAi as a Tool to Modulate Gene Expression
  • RNA interference refers to the process of sequence-specific post-transcriptional gene silencing mediated by double-stranded RNA (dsRNA). After the discovery of the phenomenon in plants in the early 1990s, Andy Fire and Craig Mello demonstrated that dsRNA specifically and selectively inhibited gene expression in an extremely efficient manner in Caenorhabditis elegans (Fire et al., 1998). The sequence of the first strand (sense RNA) coincided with that of the corresponding region of the target messenger RNA (mRNA). The second strand (antisense RNA) was complementary to the mRNA. The resulting dsRNA turned out to be several orders of magnitude more efficient than the corresponding single-stranded RNA molecules (in particular, antisense RNA).
  • The process of RNAi begins when the enzyme DICER encounters dsRNA and chops it into pieces called small-interfering RNAs or siRNA. This protein belongs to the RNase III nuclease family. A complex of proteins gathers up these siRNAs and uses their code as a guide to search out and destroy any RNAs in the cell with a matching sequence, such as target mRNA (see Bosher & Labouesse, 2000; and Akashi et al., 2001).
  • In attempting to apply RNAi for gene knockdown, it was recognized that mammalian cells have developed various protective mechanisms against viral infections that could impede the use of this approach. Indeed, the presence of extremely low levels of viral dsRNA triggers an interferon response, resulting in a global non-specific suppression of translation, which in turn triggers apoptosis (Williams, 1997, Gil & Esteban, 2000).
  • In 2000, dsRNA was reported to specifically inhibit three genes in the mouse oocyte and early embryo. Translational arrest, and thus a PKR response, was not observed as the embryos continued to develop (Wianny & Zernicka-Goetz, 2000). Research at Ribopharma AG (Kulmbach, Germany) demonstrated the functionality of RNAi in mammalian cells, using short (20-24 base pairs) dsRNAs to switch off genes in human cells without initiating the acute-phase response. Similar experiments carried out by other research groups confirmed these results (Elbashir et al., 2001; Caplen et al., 2001). Tested in a variety of normal and cancer human and mouse cell lines, it was determined that short hairpin RNAs (shRNAs) can silence genes as efficiently as their siRNA counterparts (Paddison et al., 2002). Recently, another group of small RNAs (21-25 base pairs) was shown to mediate downregulation of gene expression. These RNAs, small temporally regulated RNAs (stRNAs), regulate timing of gene expression during development in Caenorhabditis elegans (for review see Banerjee & Slack, 2002 and Grosshans & Slack, 2002).
  • Scientists have used RNAi in several systems, including Caenorhabditis elegans, Drosophila, trypanosomes, and other invertebrates. Several groups have recently presented the specific suppression of protein biosynthesis in different mammalian cell lines (specifically in HeLa cells) demonstrating that RNAi is a broadly applicable method for gene silencing in vitro. Based on these results, RNAi has rapidly become a well recognized tool for validating (identifying and assigning) gene functions. RNAi employing short dsRNA oligonucleotides will yield an understanding of the function of genes being only partially sequenced.
  • Recently, Krutzfeldt and colleagues have shown that a class of specially engineered compounds called ‘antagomirs’ can effectively silence the action of microRNAs (miRNAs), non-coding pieces of RNA that regulate gene expression (Krutzfeldt et al., 2005).
  • The preceding is a discussion of relevant art pertaining to RNAi. The discussion is provided only for understanding of the invention that follows, and is not an admission that any of the work described is prior art to the claimed invention.
  • Interleukin-12 and Crohn's Disease.
  • Interleukin-12 (1L-12) is a heterodimeric 70 kDa glycoprotein (IL12-p70) consisting of a 40 kDa subunit (designated IL12-p40) and a 35 kDa subunit (designated IL12p35) linked by disulfide bonds that are essential for the biological activity of IL-12.
  • IL-12 is a key cytokine that regulates cell-mediated immune responses and type 1 helper T (Th1) cells inflammatory reaction (Gately et al., 1998; Trinchieri, 1998). The ability of IL-12 to strongly promote the development of Th1 cells makes it an ideal target for the treatment of Th1 cell-mediated diseases, such as autoimmune diseases and inflammatory bowel disease (IBD).
  • One particular IBD is Crohn's disease, a pathology characterized by an increased production of IL-12 by antigen-presenting cells in intestinal tissue and interferon-γ and tumour necrosis factor α (TNF-α) by intestinal lymphocytes and macrophages (Fais et al., 1994; Fuss et al., 1996; Monteleone et al., 1997; Parronchi et al., 1997; Plevy et al., 1997).
  • Crohn's disease causes inflammation in the small intestine. The inflammation can cause pain and can make the intestines empty frequently, resulting in diarrhoea. The most common symptoms of Crohn's disease are abdominal pain and diarrhoea, although rectal bleeding, weight loss and fever may also occur. Bleeding may be serious and persistent, leading to anaemia. Children with Crohn's disease may suffer delayed development and stunted growth.
  • Most people are first treated with drugs containing mesalamine, a substance that helps control inflammation. Sulfasalazine is the most commonly used of these drugs. Patients who do not benefit from it or who cannot tolerate it may be put on other mesalamine-containing drugs, generally known as 5-ASA agents, such as Asacol, Dipentum or Pentasa. Possible side effects of mesalamine preparations include nausea, vomiting, diarrhoea and headache. Some patients take corticosteroids to control inflammation. These drugs are the most effective for active Crohn's disease, but they can cause serious side effects, including greater susceptibility to infection. Drugs that suppress the immune system are also used to treat Crohn's disease. Most commonly prescribed are 6-mercaptopurine and a related drug, azathioprine. Immunosuppressive agents work by blocking the immune reaction that contributes to inflammation. These drugs may cause side effects like nausea, vomiting, and diarrhoea and may lower a person's resistance to infection. Surgery to remove part of the intestine can help Crohn's disease but cannot cure it. Due to the side effects and the lack of effectiveness of the current treatments for Crohn's disease, researchers continue to look for more effective treatments.
  • Inhibiting the action of IL-12 has been shown to suppress development and clinical progression of disease in a multitude of experimental models of autoimmunity and chronic inflammation (Caspi, 1998). These models include experimental autoimmune encephalomyelitis (EAE), experimental autoimmune uveitis (EAU), collagen-induced arthritis (CIA), autoimmune nephritis, insulin-dependent diabetes mellitus (IDDM) and different models for IBD (Vandenbroeck et al., 2004). In these models, the role of endogenous IL-12 has been addressed by using IL-12p40 knockout mice or by administering anti-IL-12 antibodies.
  • In particular, targeting IL-12 with antibodies is an effective treatment for the intestinal inflammation in animal models of Crohn's disease (Mannon et al., 2004). Thus, mice with trinitrobenzene sulfonate-induced colitis have a Th1-mediated gut inflammation characterized by greatly increased production of IL-12, interferon-γ and tumour necrosis factor α (TNF-α). In mice, administration of a monoclonal antibody against IL-12 can result in the resolution of established colitis and, if given at the time of induction of colitis, can prevent inflammation (Neurath et al., 1995).
  • Anti-interleukin-12 can also prevent and treat the spontaneous colitis seen in models of Th1-mediated inflammation such as mice that over-express the human CD3e gene and mice deficient in interleukin-10 (Davidson et al., 1998; Simpson et al., 1998).
  • Data from an early phase 2 study provide some evidence that treatment with a monoclonal antibody against IL-12 p40 may induce clinical response and remission in patients with active Crohn's disease (Mannon et al., 2004). This treatment is associated with decreases in Th1-mediated inflammatory cytokines at the site of disease.
  • Previous evidence obtained from animal models, as well as the clinical effects of anti-IL-12 in patients with Crohn's disease (Mannon et al., 2004), highlight the importance of IL-12 as a target for future treatments for Crohn's disease.
  • Modulation of IL-12 Levels by Means of siRNA.
  • siRNA targeting of IL-12 expression has already been used to obtain modified dendritic cells (DC) that might be used in a variety of therapeutic in vitro, ex vivo and in vivo methods to modulate T cell activity, and thus have use in therapeutic approaches for the treatment of immune disorders in a mammalian subject (WO 03/104455; Hill et al., 2003). siRNA targeting of IL-12 expression in mature DC has revealed a critical role for IL-12 in natural killer cell interferon γ (IFN-γ) secretion promoted by mature DC (Borg et al., 2004). Further, IL-12 p35 inhibitors including siRNA have surprisingly demonstrated to block differentiation of preadipocytes to adipocytes and triglyceride accumulation in adipocytes (WO 03/104495).
  • siRNA targeting IL-12p40 has successfully been delivered by means of liposome encapsulation to murine peritoneal cavity to modulate the local and systemic inflammatory response after endotoxin challenge (Flynn et al., 2004). However, to our knowledge, there is no previous evidence of intrarectal administration of siRNA for the downregulation of IL-12 nor of any other gene involved in intestinal pathologies. We have developed techniques for downregulation of IL-12 expression in vivo to treat intestinal disorders; and we have also developed techniques for targeting siRNAs to the intestine by intrarectal administration.
  • SUMMARY OF THE INVENTION
  • The present invention provides methods and compositions for the treatment of intestinal pathologies by means of intrarectal administration of the RNAi technology. The compositions of the invention comprise short interfering nucleic acid molecules (siNA) and related compounds including, but not limited to, siRNA. In particular, the compositions of the invention can be used in the preparation of a medicament for the treatment of intestinal pathologies including: hyperproliferative diseases, in particular, colorectal cancer; autoimmune and inflammatory bowel diseases (1BD), in particular Crohn's disease; colitis, in particular ulcerative colitis; irritable bowel syndrome; infectious diseases of the intestine, such as pseudomembranous colitis, amebiasis or intestinal tuberculosis; colonic polyps; diverticular disease; constipation; intestinal obstruction; malabsorption syndromes; rectal diseases and diarrhoea. The present invention encompasses compositions and methods of use of siNA including, but not limited to, short interfering RNA (siRNA), double-stranded RNA (dsRNA), micro-RNA (miRNA), antagomirs and short hairpin RNA (shRNA) molecules capable of mediating RNA interference.
  • The methods of the invention comprise the administration to a patient in the need thereof of an effective amount of one or more siNA of the invention for the treatment of an intestinal condition. In preferred embodiments, the methods of the invention comprise intrarectal administration of the therapeutic siNA.
  • In one embodiment, the present invention relates to siNA or similar chemically synthesized entities that are directed at interfering with the mRNA expression of either the p35 or the p40 subunits of the cytokine IL-12, and that ultimately modulate the amount of protein produced. Compositions and methods comprising above-mentioned siRNA and related compounds are intended for the treatment of diseases associated with over-expression of IL-12, such as autoimmune diseases and inflammatory bowel diseases (IBD), in particular, Crohn's disease.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Oligonucleotide sequences for siRNA molecules targeting IL-12 p35 and p40 subunits encompassed by the present invention. The SEQ ID Nos given in the Figure refer to the sense (5′->3′) strand; typically sIRNA will be administered as dsRNA, so will include both the sense strand and its complement.
  • FIG. 2. Effect of siRNA on IL-12 p35 subunit expression in an in vitro system. siRNA treatment reduces the levels of IL-12 p35 gene transcript. RNA was prepared from SW480 cells treated with siRNAs for different times. The samples were analyzed by RT-PCR using specific primers. The values show the mean expression levels of different transcripts normalized to 18S as housekeeping gene.
  • FIG. 3. Effect of siRNA on IL-12 p40 subunit expression in an in vitro system. A: siRNA treatment reduces the levels of IL-12 p40 gene transcript in human cells. RNA was prepared from SW480 cells treated with siRNA SEQ ID 67 and SEQ ID 79 at different times, at dose treatment of 200 nM. The values show the mean expression levels of different transcripts normalized to 18S as housekeeping gene. The values represent the mean of the percentage of the normalized mRNA levels upon siRNA interference over the control gene expression and their medium standard deviations (SEM). B: siRNA treatment reduces the levels of IL-12 p40 gene transcript in murine cells. RNA was prepared from C2C12 cells treated with siRNA SEQ ID 86 and SEQ ID 87 at different times, at dose treatment of 100 nM. SEQ ID 86, which is homologous to human SEQ ID 67, targets the mouse IL-12 p40 subunit. Further targeting the mouse IL-12 p40 subunit, SEQ ID 87 is the siRNA with the best score in mouse, and has no homologous siRNA duplex in human. siNA molecules SEQ ID 86 and SEQ ID 87 are as described below, with 2 thymidine nucleotide 3′ overhangs. The values represent the mean of the percentage of the normalized mRNA levels compared to 18S upon siRNA interference over the control gene expression and their medium standard deviations (SEM).
  • FIG. 4. siRNA treatment reduces the levels of GFP gene transcript in small intestine. The collected tissue in OCT was analyzed by microscopy and measured by photoshop program. Data show single dose siRNA treatment (mice 2-3) and repeated dose treatment (mice 4-5). The values show the expression levels of 25 representative images per mouse referred to control untreated mouse. Standard deviation of the data is represented.
  • FIG. 5. siRNA treatment reduces the levels of GFP gene transcript in small intestine. The tissue collected in RNA later was analyzed by RT-PCR. Data show single dose siRNA treatment (mice 2-3) and repeated dose treatment (mice 4-5). Standard deviation is represented.
  • FIG. 6. siRNA treatment reduces the levels of GFP gene transcript in large intestine. The tissue collected in OCT was analyzed by microscopy and measured by photoshop program. Data show single dose siRNA treatment (mice 2-3) and repeated dose treatment (mice 4-5). The values show the expression levels of 25 representative images per mouse referred to control untreated mouse. Standard deviation of the data is represented.
  • FIG. 7: siRNA treatment reduces the levels of GFP gene transcript in large intestine. The collected tissue in RNA later was analyzed by RT-PCR. Data show single dose siRNA treatment (mice 2-3) and repeated dose treatment (mice 4-5). Standard deviation is represented.
  • FIG. 8: Data of samples collected in OCT medium.
  • FIG. 9: Data of samples collected in RNA later.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to methods and compositions for the treatment of intestinal pathologies by means of intrarectal administration of the RNAi technology. The compositions of the invention comprise short interfering nucleic acid molecules (siNA) that modulate the expression of target genes associated with conditions of the intestinal wall.
  • The methods of the invention comprise the administration to a patient in need thereof of an effective amount of one or more siNA of the invention.
  • Design of siRNA
  • A gene is “targeted” by siNA according to the invention when, for example, the siNA selectively decrease or inhibit the expression of the gene. Alternatively, siNA target a gene when the siNA hybridize under stringent conditions to the gene transcript. siNA can be tested either in vitro or in vivo for the ability to target a gene.
  • In 1999, Tuschl et al. deciphered the silencing effect of siRNAs showing that their efficiency is a function of the length of the duplex, the length of the 3′-end overhangs, and the sequence in these overhangs.
  • Selecting the right homologous region within the target gene is of great relevance for accurate silencing. A short fragment of the target gene sequence (e.g., 19-40 nucleotides in length) is chosen as the sequence of the siNA of the invention. In one embodiment, the siNA is siRNA. In such embodiments, the short fragment of target gene sequence is a fragment of the target gene mRNA. In preferred embodiments, the criteria for choosing a sequence fragment from the target gene mRNA to be a candidate siRNA molecule include: 1) a sequence from the target gene mRNA that is at least 50-100 nucleotides from the 5′ or 3′ end of the native mRNA molecule; 2) a sequence from the target gene mRNA that has a G/C content of between 30% and 70%, most preferably around 50%; 3) a sequence from the target gene mRNA that does not contain repetitive sequences (e.g., AAA, CCC, GGG, TTT, AAAA, CCCC, GGGG, TTTT); 4) a sequence from the target gene mRNA that is accessible in the mRNA; and 5) a sequence from the target gene mRNA that is unique to the target gene. The sequence fragment from the target gene mRNA may meet one or more of the above-mentioned identified criteria. In preferred embodiments, the siRNA has a G/C content below 60% and/or lacks repetitive sequences.
  • Practically, the gene of interest is introduced as a nucleotide sequence in a prediction program that takes into account all the variables described above for the design of optimal oligonucleotides. This program scans any mRNA nucleotide sequence for regions susceptible to be targeted by siRNA. The output of this analysis is a score of possible siRNA oligonucleotides. The highest scores are used to design double stranded RNA oligonucleotides (typically 21 bp long, although other lengths are also possible) that are typically made by chemical synthesis. We plan to test several chemical modifications that are well known in the art. These modifications are aimed at increasing stability or availability of the dsRNA oligonucleotides.
  • Candidate oligonucleotides can further be filtered for interspecies sequence conservation in order to facilitate the transition from animal to human clinical studies.
  • In addition to siNA which is perfectly complementary to the target region, degenerate siNA sequences may be used to target homologous regions. WO2005/045037 describes the design of siNA molecules to target such homologous sequences, for example by incorporating non-canonical base pairs, for example mismatches and/or wobble base pairs, that can provide additional target sequences. In instances where mismatches are identified, non-canonical base pairs (for example, mismatches and/or wobble bases) can be used to generate siNA molecules that target more than one gene sequence. In a non-limiting example, non-canonical base pairs such as UU and CC base pairs are used to generate siNA molecules that are capable of targeting sequences for differing targets that share sequence homology. As such, one advantage of using siNAs of the invention is that a single siNA can be designed to include nucleic acid sequence that is complementary to the nucleotide sequence that is conserved between homologous genes. In this approach, a single siNA can be used to inhibit expression of more than one gene instead of using more than one siNA molecule to target different genes.
  • Sequence identity may be calculated by sequence comparison and alignment algorithms known in the art (see Gribskov and Devereux, Sequence Analysis Primer, Stockton Press, 1991, and references cited therein) and calculating the percent difference between the nucleotide sequences by, for example, the Smith-Waterman algorithm as implemented in the BESTFIT software program using default parameters (e.g., University of Wisconsin Genetic Computing Group). Greater than 90%, 95%, or 99% sequence identity between the siNA and the portion of the target gene is preferred. Alternatively, the complementarity between the siNA and native RNA molecule may be defined functionally by hybridisation as well as functionally by its ability to decrease or inhibit the expression of a target gene. The ability of a siNA to affect gene expression can be determined empirically either in vivo or in vitro.
  • Preferred siNA molecules of the invention are double stranded. In one embodiment, double stranded siNA molecules comprise blunt ends. In another embodiment, double stranded siNA molecules comprise overhanging nucleotides (e.g., 1-5 nucleotide overhangs, preferably 2 nucleotide overhangs). In a specific embodiment, the overhanging nucleotides are 3′ overhangs. In another specific embodiment, the overhanging nucleotides are 5′ overhangs. Any type of nucleotide can be a part of the overhang. In one embodiment, the overhanging nucleotide or nucleotides are ribonucleic acids. In another embodiment, the overhanging nucleotide or nucleotides are deoxyribonucleic acids. In a preferred embodiment, the overhanging nucleotide or nucleotides are thymidine nucleotides. In another embodiment, the overhanging nucleotide or nucleotides are modified or non-classical nucleotides. The overhanging nucleotide or nucleotides may have non-classical internucleotide bonds (e.g., other than phosphodiester bond).
  • Synthesis of siNA Duplexes
  • siNA can be synthesized by any method known in the art. RNAs are preferably chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer. Additionally, siRNA can be obtained from commercial RNA oligo synthesis suppliers, including, but not limited to, Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, Colo., USA), Glen Research (Sterling, Va., USA), ChemGenes (Ashland, Mass., USA), and Cruachem (Glasgow, UK), Qiagen (Germany), Ambion (USA) and Invitrogen (Scotland). Alternatively, siNA molecules of the invention can be expressed in cells by transfecting the cells with vectors containing the reverse complement siNA sequence under the control of a promoter. Once expressed, the siNA can be isolated from the cell using techniques well known in the art.
  • An annealing step is necessary when working with single-stranded RNA molecules. To anneal the RNAs, 30 μl of each RNA oligo 50 μM solution are to be combined in 100 mM potassium acetate, 30 mM HEPES-KOH pH 7.4, 2 mM magnesium acetate. The solution is then incubated for 1 minute at 90° C., centrifuged for 15 seconds, and incubated for 1 hour at 37° C.
  • In embodiments where the siRNA is a short hairpin RNA (shRNA); the two strands of the siRNA molecule may be connected by a linker region (e.g., a nucleotide linker or a non-nucleotide linker).
  • Chemical Modification of siNA.
  • The siNAs of the invention may contain one or more modified nucleotides and/or non-phosphodiester linkages. Chemical modifications well known in the art are capable of increasing stability, availability, and/or cell uptake of the siNA. The skilled person will be aware of other types of chemical modification which may be incorporated into RNA molecules (see International Publications WO03/070744 and WO2005/045037 for an overview of types of modifications).
  • In one embodiment, modifications can be used to provide improved resistance to degradation or improved uptake. Examples of such modifications include phosphorothioate internucleotide linkages, 2′-O-methyl ribonucleotides (especially on the sense strand of a double stranded siRNA), 2′-deoxy-fluoro ribonucleotides, 2′-deoxy ribonucleotides, “universal base” nucleotides, 5-C-methyl nucleotides, and inverted deoxyabasic residue incorporation (see generally GB2406568).
  • In another embodiment, modifications can be used to enhance the stability of the siRNA or to increase targeting efficiency. Modifications include chemical cross linking between the two complementary strands of an siRNA, chemical modification of a 3′ or 5′ terminus of a strand of an siRNA, sugar modifications, nucleobase modifications and/or backbone modifications, 2′-fluoro modified ribonucleotides and 2′-deoxy ribonucleotides (see generally International Publication WO2004/029212).
  • In another embodiment, modifications can be used to increase or decrease affinity for the complementary nucleotides in the target mRNA and/or in the complementary siNA strand (see generally International Publication WO2005/044976). For example, an unmodified pyrimidine nucleotide can be substituted for a 2-thio, 5-alkynyl, 5-methyl, or 5-propynyl pyrimidine. Additionally, an unmodified purine can be substituted with a 7-deza, 7-alkyl, or 7-alkenyl purine.
  • In another embodiment, when the siNA is a double-stranded siRNA, the 3′-terminal nucleotide overhanging nucleotides are replaced by deoxyribonucleotides (see generally Elbashir et al., 2001).
  • In Vitro Testing of siRNA Duplexes.
  • To check the specificity of the siRNA interference, cell cultures expressing the target genes were employed.
  • In the case of IL-12 p35 and p40 subunits, the cells used for the experiments were human SW480 cells and murine muscle cells C2C12. After incubation of the cells with the corresponding siRNA duplexes, the levels of p35 and p40 expression were analyzed. For linking siRNA knockdown to specific phenotypes in cultured cells, it is necessary to demonstrate the decrease of the targeted protein or at least to demonstrate the reduction of the targeted mRNA.
  • mRNA levels of the target gene can be quantitated by real time PCR(RT-PCR). Further, the protein levels can be determined in a variety of ways well known in the art, such as Western blot analysis with specific antibodies to the different target allow direct monitoring of the reduction of targeted protein.
  • siRNA are introduced into cells by means of any transfection technique well known in the art. A single transfection of siRNA duplex can be performed, for instance, by using a cationic lipid, such as Lipofectamine 2000 Reagent (Invitrogen), followed by an assay of silencing efficiency 24, 48 and 72 hours after transfection.
  • A typical transfection protocol can be performed as follows: for one well of a E-well plate, we transfect using 100 nM for murine C2C12 cells or 200 nM for human SW480 cells as final concentration of siRNA. Following Lipofectamine 2000 Reagent protocol, the day before transfection, we seed 2-4×105 cells per well in 3 ml of an appropriate growth medium, containing DMEM, 10% serum, antibiotics and glutamine, and incubate cells under normal growth conditions (37° C. and 5% CO2). On the day of transfection, cells have to be at 30-50% confluence. We dilute 12.5 μl of 20 μM siRNA duplex (corresponding to 100 nM final concentration) or 25 μl of 20 μM siRNA duplex (corresponding to 200 nM final concentration) in 250 μl of DMEM and mix. Also, 6 μl of Lipofectamine 2000 is diluted in 250 μl of DMEM and mixed. After a 5 minute incubation at room temperature, the diluted oligomer (siRNA duplex) and the diluted Lipofectamine are combined to allow complex formation during a 20 minutes incubation at room temperature. Afterwards, we add the complexes drop-wise onto the cells with 2 ml of fresh growth medium low in antibiotics and mix gently by rocking the plate back and forth, to ensure uniform distribution of the transfection complexes. We incubate the cells under their normal growth conditions and the day after, the complexes are removed and fresh and complete growth medium is added. To monitor gene silencing, cells are collected at 24, 48 and 72 h post-transfection.
  • The efficiency of transfection may depend on the cell type, but also on the passage number and the confluency of the cells. The time and the manner of formation of siRNA-liposome complexes (e.g. inversion versus vortexing) are also critical. Low transfection efficiencies are the most frequent cause of unsuccessful silencing. Good transfection is a non-trivial issue and needs to be carefully examined for each new cell line to be used. Transfection efficiency may be tested transfecting reporter genes, for example a CMV-driven EGFP-expression plasmid (e.g. from Clontech) or a B-Gal expression plasmid, and then assessed by phase contrast and/or fluorescence microscopy the next day.
  • Depending on the abundance and the life time (or turnover) of the targeted protein, a knock-down phenotype may become apparent after 1 to 3 days, or even later. In cases where no phenotype is observed, depletion of the protein may be observed by immunofluorescence or Western blotting.
  • After transfections, total RNA fractions extracted from cells are pre-treated with DNase I and used for reverse transcription using a random primer. PCR-amplified with a specific primer pair covering at least one exon-exon junction is used as control for amplification of pre-mRNAs. RT-PCR of a non-targeted mRNA is also needed as control. Effective depletion of the mRNA yet undetectable reduction of target protein may indicate that a large reservoir of stable protein may exist in the cell. Alternatively, RT-PCR amplification can be used to test in a more precise way the mRNA decrease or disappearance. RT-PCR quantitates the initial amount of the template most specifically, sensitively and reproducibly. RT-PCR monitors the fluorescence emitted during the reaction as an indicator of amplicon production during each PCR cycle, in a light cycler apparatus. This signal increases in direct proportion to the amount of PCR product in a reaction. By recording the amount of fluorescence emission at each cycle, it is possible to monitor the PCR reaction during exponential phase where the first significant increase in the amount of PCR product correlates to the initial amount of target template.
  • To verify the interference pattern of the differentially expressed IL-p35 and p40 genes in the cell cultures, quantitative RT-PCR was performed. For quantitative RT-PCR, approximately 500 ng of total RNA was used for reverse transcription, followed by PCR amplification with specific primers for each gene in reaction. The PCR conditions were an initial step of 30 s at 95° C., followed by 40 cycles of 5 s at 95° C., 10 s at 62° C. and 15 s at 72° C. Quantification of 18S mRNA was used as a housekeeping gene as a control for data normalization. Relative gene expression comparisons work best when the gene expression of the chosen endogenous/internal control is more abundant and remains constant, in proportion to total RNA, among the samples. By using an invariant endogenous control as an active reference, quantitation of an mRNA target can be normalised for differences in the amount of total RNA added to each reaction. The amplification curves obtained with the light cycler were analyzed in combination with the control kit DNA, which targets in vitro transcribed beta-globin DNA template, according to the manufacturer protocol. In order to assess the specificity of the amplified PCR product a melting curve analysis was performed. The resulting melting curves allow discrimination between primer-dimers and specific PCR product.
  • Intrarectal Administration of siNA.
  • Intrarectal siNA delivery studies were carried out in GFP C57BL/6-TG (ACTB-EGFP) mice. This transgenic mouse line was bought from “The Jackson Laboratory”. Transgenic mice have been used because homozygous mice for this transgene die within the first two weeks following birth. The transgenic mouse line with an “enhanced” GFP (EGFP) cDNA under the control of a chicken beta-actin promoter and cytomegalovirus enhancer makes all of the tissues, with the exception of erythrocytes and hair, appear green under excitation light. This strain was generated in C57BL/6 mice. The strain cDNA encoding enhanced green fluorescent protein (EGFP) was adjoined to the chicken beta actin promoter and cytomegalovirus enhancer. A bovine globin polyadenylation signal was also included in the construct. The EcoR1 sites included in the PCR primers were used to introduce the amplified EGFP cDNA into a pCAGGS expression vector containing the chicken beta-actin promoter and cytomegalovirus enhancer, beta-actin intron and bovine globin poly-adenylation signal. The entire insert with the promoter and coding sequence was excised with Bam-HI and SalI and gel-purified.
  • The siRNA duplex used for intrarectal injection in mice was purchased from Dharmacon. Dharmacon Research Inc (Lafayette, Colo.) have developed a new generation of modified siRNA for in vivo use as a therapeutic, named siSTABLEv2. Dharmacon's siSTABLEv2 siRNA have demonstrated an enhanced stability in serum with respect to that of non-modified siRNA. Conventional siRNA are typically degraded within minutes in serum-containing environments, making in vivo use of siRNA problematic. The siSTABLEv2 modification dramatically extends the siRNA stability in serum as described in Dharmacon's web page (http://www.dharmacon.com/docs/siSTABLE %20v2%20Flier.pdf).
  • The siRNA used to downregulate EGFP mRNA expression targeted the following sequence in EGFP mRNA: 5′-GGC UAC GUC CAG GAG CGC ACC-3′ (SEQ ID No 88). The sense strand of the siRNA duplex was 5′-P GGC UAC GUC CAG CGC ACC-3′ (SEQ ID No 89) and the antisense strand was 5′-P U GCG CUC CUG GAC GUA GCC UU-3′ (SEQ ID No 90). This sequence is distributed by Dharmacon as pre-synthesized control siRNA green fluorescent protein duplex.
  • For the intrarectal delivery experiments C57BL/6-TG (ACTB-EGFP) mice (males, 8 weeks old) were used. The animals were kept in cages with free access to food and water until one day before the experimental protocol. For intrarectal therapeutic silencing, mice were fasted for one day prior to the treatment. The drugs are typically administered by injecting a small volume (120 μL) in the rectum. Control mouse is treated with the vehicle alone. In all cases animals were sacrificed two days after the first injection by cervical dislocation. The protocol for the siRNA application in mouse is as follows. For each experimental administration, 60 μl siRNA duplexes were premixed with 60 μl of NaCl (1.8% w/v) up to physiological levels. In all cases animals were sacrificed two days after the first injection.
  • Experimental conditions used are indicated in the Table below. Each condition was analyzed in duplicate. Mice 2 and 3 were treated intrarectally with one dose of 250 μg (19 nanomols) of the siRNA vs GFP, while mice 4 and 5 were treated with two doses of 125 μg of siRNA during two consecutive days.
  • TABLE
    Schematic distribution of experimental conditions
    for intrarectal siRNA delivery.
    Mouse number Intrarectal Therapeutic Treatment
    1 Vehicle control dose
    2 Single dose of 250 μg of siRNA
    3 Single dose of 250 μg of siRNA
    4 Two doses of 125 μg of siRNA each 24 h
    5 Two doses of 125 μg of siRNA each 24 h
    Doses of siRNA are indicated in the table.
  • The sample tissues were collected and analyzed by two methods: One in OCT medium and another in RNA later (Ambion). OCT blocks were storaged at −80° C. until data processing. OCT blocks were cut in slices of 12 μm by a cryostat (Leica CM 1850) at −20° C. The collected slices were analyzed on a fluorescence microscope (Olympus BX51) coupled to a digital camera (DP70), using a filter of 488 nm. The sensitive conditions (ISO200), resolution image size (2040×1536) and time exposure (1 second) were set up for all the samples in order to be compared between them. Green fluorescence was measured as an index of GFP expression by an Adobe Photoshop program (version 8.0). By this method, 25 different data were collected for each analyzed tissue. Tissues isolated in RNA later were stored at −20° C. RNA later was removed before RNA extraction. RNA was isolated with the Trizol Reagent (Invitrogen) according to the manufacturer protocol. DNAse treatment was done before measurement of GFP expression by RT-PCR as described above.
  • Pharmaceutical Formulations and Routes of Administration.
  • The present invention may comprise the administration of one or more species of siNA molecule simultaneously. These species may be selected to target one or more target genes.
  • In one embodiment, a single type of siNA is administered in the therapeutic methods of the invention. In another embodiment, a siNA of the invention is administered in combination with another siNA of the invention and/or with one or more other non-siNA therapeutic agents useful in the treatment, prevention or management of a disease condition of the intestine wall. The term “in combination with” is not limited to the administration of therapeutic agents at exactly the same time, but rather it is meant that the siNAs of the invention and the other agent are administered to a patient in a sequence and within a time interval such that the benefit of the combination is greater than the benefit if they were administered otherwise. For example, each therapeutic agent may be administered at the same time or sequentially in any order at different points in time; however, if not administered at the same time, they should be administered sufficiently close in time so as to provide the desired therapeutic effect. Each therapeutic agent can be administered separately, in any appropriate form and by any suitable route.
  • The siNAs of the invention may be formulated into pharmaceutical compositions by any of the conventional techniques known in the art (see for example, Alfonso, G. et al., 1995, in: The Science and Practice of Pharmacy, Mack Publishing, Easton Pa., 19th ed.). Formulations comprising one or more siNAs for use in the methods of the invention may be in numerous forms, and may depend on the various factors specific for each patient (e.g., the type and severity of disorder, type of siNA administered, age, body weight, response, and the past medical history of the patient), the number and type of siNAs in the formulation, the form of the composition (e.g., in liquid, semi-liquid or solid form), the therapeutic regime (e.g. whether the therapeutic agent is administered over time as a slow infusion, a single bolus, once daily, several times a day or once every few days), and/or the route of administration (e.g., topical, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, or sublingual means).
  • The siNA molecules of the invention and formulations or compositions thereof may be administered directly or topically as is generally known in the art. For example, a siNA molecule can comprise a delivery vehicle, including liposomes, for administration to a subject. Carriers and diluents and their salts can be present in pharmaceutically acceptable formulations. 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 poly (lactic-co-glycolic) acid (PLGA) and PLCA microspheres, biodegradable nanocapsules, and bioadhesive microspheres, or by proteinaceous vectors. In another embodiment, the nucleic acid 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.
  • A siNA molecule of the invention may be complexed with membrane disruptive agents and/or a cationic lipid or helper lipid molecule.
  • Delivery systems which may be used with the invention include, for example, aqueous and non aqueous gels, creams, multiple emulsions, microemulsions, liposomes, ointments, aqueous and non aqueous solutions, lotions, aerosols, hydrocarbon bases and powders, and can contain excipients such as solubilizers, permeation enhancers (e.g., fatty acids, fatty acid esters, fatty alcohols and amino acids), and hydrophilic polymers (e.g., polycarbophil and polyvinylpyrolidone). In one embodiment, the pharmaceutically acceptable carrier is a liposome or a transdermal enhancer.
  • A pharmaceutical formulation of the invention is 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, or by injection. 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.
  • The present invention also includes compositions prepared for storage or administration that include a pharmaceutically effective amount of the desired compounds in a pharmaceutically acceptable carrier or diluent. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art. For example, preservatives, stabilizers, dyes and flavouring agents can be provided. These include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid. In addition, antioxidants and suspending agents can be used.
  • A pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, preferably all of the symptoms) of a disease state. The pharmaceutically effective dose depends on the type of disease, the composition used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors that those skilled in the medical arts will recognize.
  • The formulations of the invention can be administered in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and/or vehicles. Formulations can be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. 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 monostearate 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.
  • 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 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 colouring agents, one or more flavouring agents, and one or more 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 anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents or suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, can also be present.
  • 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 flavouring 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.
  • A sterile injectable preparation can also 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.
  • 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.
  • 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 anaesthetics, preservatives and buffering agents can be dissolved in the vehicle.
  • For administration to non-human animals, the composition can also be added to the animal feed or drinking water. It can be convenient to formulate the animal feed and drinking water compositions so that the animal takes in a therapeutically appropriate quantity of the composition along with its diet. It can also be convenient to present the composition as a premix for addition to the feed or drinking water.
  • The nucleic acid molecules of the present invention can also be administered to a subject in combination with other therapeutic compounds to increase the overall therapeutic effect. The use of multiple compounds to treat an indication can increase the beneficial effects while reducing the presence of side effects.
  • Alternatively, certain siNA molecules of the invention can be expressed within cells from eukaryotic promoters. Recombinant vectors capable of expressing the siNA molecules can be delivered and persist in target cells. Alternatively, vectors can be used that provide for transient expression of nucleic acid molecules. Such vectors can be repeatedly administered as necessary. Once expressed, the siNA molecule interacts with the target mRNA and generates an RNAi response. Delivery of siNA molecule expressing vectors can be systemic, such as by intravenous or intramuscular 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.
  • Results.
  • Example 1 Design of siNA
  • GenBank Accession numbers corresponding to IL-12 p35 (Interleukin 12A, natural killer cell stimulatory factor 1, cytotoxic lymphocyte maturation factor 1, p35) and p40 (Interleukin 12B, natural killer cell stimulatory factor 2, cytotoxic lymphocyte maturation factor 2, p40) subunits are NM 000882 and NM 002187, respectively.
  • Corresponding mRNA nucleotide sequences were introduced within the proprietary prediction program described above, and siNA molecules directed to target IL-12 p35 and p40 subunits were obtained. The output of this analysis was a score of possible siNA oligonucleotides, the highest scores being used to design double stranded RNA oligonucleotides (typically 19 bp long) that were typically made by chemical synthesis.
  • In preferred embodiments, siNA compositions of the invention are any of SEQ ID NOS:1-81 of FIG. 1; typically administered as a duplex of the sense strand and the antisense strand. The invention also encompasses siNA that are 40 nucleotides or less and comprise a nucleotide sequence of any of SEQ ID NOS:1-81. In a specific embodiment, the siNA is 21-30 nucleotides long and comprises any one of SEQ ID NOS:1-81 of FIG. 1. All siNA molecules used in the experiments described below were designed to have a 2 thymidine nucleotide 3′ overhang.
  • Example 2 In Vitro Assays for IL-12 p35
  • To determine the inhibition of the IL-12 p35 target gene, a panel of siRNA contained within FIG. 1 has been analysed in cell cultures. SiRNA with the best characteristics were selected to be tested and were applied to proper cell cultures, such as SW480. The effect of siRNA over the target gene was analyzed by RT-PCR according to the manufacturer's protocol. The gene target transcript levels were normalized using 18S as housekeeping gene. Some of the different siRNA that were tested and their different efficacies in the interference of the target gene are included in the FIG. 2. These results correspond to SEQ ID 8 and SEQ ID 17 of FIG. 1 in SW480 cells expressing p35. The values represent the mean of the percentage of the normalized mRNA levels upon siRNA interference over the control gene expression and their medium standard deviations (SEM). The level of the p35 transcript after the siRNA treatment was highly reduced with the siRNAs corresponding to SEQ ID 8 and SEQ ID 17, in SW480 compared to the control cells. The decrease of the gene expression depends on the efficiency in siRNA silencing. In fact, siRNA SEQ ID 8 treatment decreased the p35 gene expression to 56% at 24 h compared to the control.
  • Example 3 In Vitro Assays for IL-12 p40
  • To determine the inhibition of the IL-12 p40 target gene, a panel of siRNA contained within FIG. 1 has been analyzed. The siRNA with the best characteristics designed as described before, were tested in human and murine cells. The p40 transcript level was analyzed by RT-PCR and normalized using 18S as housekeeping gene. These results correspond to SEQ ID 67, SEQ ID 79 in SW480 cells expressing p40 (FIG. 3A); and SEQ ID 86 and SEQ ID 87 in C2C12 cells expressing p40 (FIG. 3B). siNA molecules SEQ ID 86 and SEQ ID 87 are as described in the figure, with 2 thymidine nucleotide 3′ overhangs.
  • The level of the p40 transcript was highly reduced after the treatment with the siRNA corresponding to SEQ ID 67 in SW480, up to 65% compared to control cells. In C2C12 cells siRNA corresponding to SEQ ID 86 decreased the gene expression to 61% at 48 h compared to the control. It is important to note that SEQ ID 67 and SEQ ID 86 correspond to homologous regions of human and mouse IL-12 p40 gene, respectively.
  • A summary of the experiments of FIGS. 2 and 3 is displayed in the following Table:
  • Gene
    Expression (%) SEM
    P35 (SW480) Control 100 0
    SEQ ID 8, 24 h 56 13.9772455
    SEQ ID 8, 48 h 73 13.6336806
    SEQ ID 17, 24 h 67 17.8860754
    SEQ ID 17, 48 h 73 20.7305043
    p40 (SW480) Control 100 0
    SEQ ID 67, 24 h 65 8.58321816
    SEQ ID 67, 48 h 86 20.4353968
    SEQ ID 79, 24 h 69 17.8276602
    SEQ ID 79, 48 h 84 13.1055338
    p40 (C2C12) Control 100 0
    SEQ ID 86, 24 h 121 13.8703694
    SEQ ID 86, 4 8h 61 23.4419624
    SEQ ID 87, 24 h 108 35.8061452
    SEQ ID 87, 48 h 85 17.1974522
  • Example 4 In Vivo Assays. Analysis of the Small Intestine
  • The siRNA application is made in order to determine the proper siRNA delivery in the intestine. To determine the siRNA effect, small intestine samples were collected in OCT medium and analyzed as previously described. Since the goal is to determine the downregulation of GFP gene transcript, levels of fluorescence were measured following siRNA application. No secondary effects were observed in the animals during the experimental protocols.
  • The first group of work (animals 2 and 3) was treated with a single dose of 250 μg of siRNA and sacrificed 48 h later. The results indicate a significant decrease of fluorescence when compared with the control mouse. Moreover, when the siRNA (250 μg) is administered in two doses of 125 μg and analyzed 48 h after the first injection, the decrease of GFP expression was similar to that after a single application. The results are shown in FIG. 4. For each experimental condition an average of the data is represented.
  • At the same time, small intestine samples were collected in RNA later to confirm previous data. mRNA levels were measured by RT-PCR. These results, shown in FIG. 5, confirm the previous ones obtained with fluorescence analysis.
  • As shown in FIG. 5, the administered dose of 250 μg of siRNA in one or two applications was enough and sufficient to downregulate the level of GFP mRNA in small intestine, confirming the delivery of the siRNA in small intestine by intrarectal administration. The level of downregulation compared to the control is higher when the analysis is done by RT-PCR, this being due to the higher sensitivity of the technique.
  • Example 5 In Vivo Assays. Analysis of Large Intestine
  • Large intestine was further analyzed in the same way as small intestine. To determine the siRNA effect in large intestine, samples collected in OCT medium were analyzed to determine the downregulation of GFP by measurement of fluorescence following siRNA application. The results indicate a significant decrease of fluorescence when compared to control mouse (FIG. 6). Moreover, when the dose is administered in two applications of 125 μg and analyzed 48 h after the first injection, the decrease was very similar to that obtained after a single siRNA administration, demonstrating the effectiveness of the treatment.
  • As in small intestine, large intestine samples were collected in RNA later and data of mRNA levels represented in FIG. 7. The data obtained by RT-PCR confirm the previous ones obtained with fluorescence analysis. These results open a new route to therapeutic siRNA administration to treatment of bowel diseases.
  • Data of samples collected in OCT medium and in RNA later are summarized in FIGS. 8 and 9 respectively.
  • We also investigated whether there was any downregulation of GFP expression in other selected tissues of the mice; no downregulation was observed in bladder, kidney, lung, ovary, and liver tissues, suggesting that intrarectal administration of siRNA can be used to specifically target intestinal tissue.
  • REFERENCES
    • Akashi H, Miyagishi M, Taira K. Suppression of gene expression by RNA interference in cultured plant cells. Antisense Nucleic Acid Drug Dev, 2001, 11(6):359-67.
    • Banerjee D, Slack F. Control of developmental timing by small temporal RNAs: a paradigm for RNA-mediated regulation of gene expression. Bioessays, 2002, 24(2):119-29.
    • Bosher J M, Labouesse M. RNA interference: genetic wand and genetic watchdog. Nat Cell Biol, 2000, 2(2):E31-6.
    • Borg C, Jalil A, Laderach D, Maruyama K, Wakasugi H, Charrier S, Ryffel B, Cambi A, Figdor C, Vainchenker W, Galy A, Caignard A, Zitvogel L. NK cell activation by dendritic cells (DCs) requires the formation of a synapse leading to 1L-12 polarization in DCs. Blood, 2004 Nov. 15; 104(10):3267-75.
    • Caplen, N. J., Parrish, S., Imani, F., Fire, A. & Morgan, R. A. Specific inhibition of gene expression by small double stranded RNAs in invertebrate and vertebrate systems. Proc. Natl. Acad. Sci. USA, 2001, 98: 9742-9747.
    • Caspi R R. IL-12 in autoimmunity. Clin Immunol Immunopathol., 1998; 88(1):4-13.
    • Davidson N J, Hudak S A, Lesley R E, Menon S, Leach M W, Rennick D M. IL-12, but not IFN-gamma, plays a major role in sustaining the chronic phase of colitis in IL-10-deficient mice. J. Immunol., 1998; 161(6):3143-9.
    • Elbashir S M, Lendeckel W, Tuschl T. RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev, 2001, 15(2):188-200.
    • Fais S, Capobianchi M R, Silvestri M, Mercuri F, Pallone F, Dianzani F. Interferon expression in Crohn's disease patients: increased interferon-gamma and -alpha mRNA in the intestinal lamina propria mononuclear cells. J Interferon Res., 1994; 14(5):235-8.
    • Fire A, Xu S, Montgomery M K, Kostas S A, Driver S E, Mello C C. Potent and specific genetic interference by double stranded RNA in Caenorhabditis elegans. Nature, 1998, 391(6669):806-11.
    • Flynn M A, Casey D G, Todryk S M, Mahon B P. Efficient delivery of small interfering RNA for inhibition of IL-12p40 expression in vivo. J Inflamm (Lond), 2004 Oct. 1; 1(1):4.
    • Fuss I J, Neurath M, Boirivant M, Klein J S, de la Motte C, Strong S A, Fiocchi C, Strober W. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn's disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J. Immunol., 1996; 157(3): 1261-70.
    • Gately M K, Renzetti L M, Magram J, Stern A S, Adorini L, Gubler U, Presky D H. The interleukin-12/interleukin-12-receptor system: role in normal and pathologic immune responses. Annu Rev Immunol, 1998; 16:495-521.
    • Gil J, Esteban M. Induction of apoptosis by the dsRNA-dependent protein kinase (PKR): mechanism of action. Apoptosis, 2000, 5(2):107-14.
    • Grosshans H, Slack F J. Micro-RNAs: small is plentiful. J Cell Biol, 2002, 156(1):17-21.
    • Hill J A, Ichim T E, Kusznieruk K P, Li M, Huang X, Yan X, Zhong R, Cairns E, Bell D A, Min W P. Immune modulation by silencing IL-12 production in dendritic cells using small interfering RNA. J. Immunol., 2003 Sep. 15; 171(6):3303.
    • Krutzfeldt J, Rajewsky N, Braich R, Rajeev K G, Tuschl T, Manoharan M, Stoffel M. Silencing of microRNAs in vivo with ‘antagomirs’. Nature 2005, 438(7068):685-9.
    • Mannon P J, Fuss I J, Mayer L, Elson C O, Sandborn W J, Present D, Dolin B, Goodman N, Groden C, Hornung R L, Quezado M, Neurath M F, Salfeld J, Veldman G M, Schwertschlag U, Strober W; Anti-1L-12 Crohn's Disease Study Group. Anti-interleukin-12 antibody for active Crohn's disease. N Engl J. Med., 2004; 351(20): 2069-79.
    • Monteleone G, Biancone L, Marasco R, Morrone G, Marasco O, Luzza F, Pallone F. Interleukin 12 is expressed and actively released by Crohn's disease intestinal lamina propria mononuclear cells. Gastroenterology, 1997; 112(4):1169-78.
    • Neurath M F, Fuss I, Kelsall B L, Stuber E, Strober W. Antibodies to interleukin 12 abrogate established experimental colitis in mice. J Exp Med., 1995; 182(5):1281-90.
    • Paddison P J, Caudy A A, Bernstein E, Hannon G J, Conklin D S. Short hairpin RNAs (shRNAs) induce sequence-specific silencing in mammalian cells. Genes Dev, 2002, 16(8):948-58.
    • Parronchi P, Romagnani P, Annunziato F, Sampognaro S, Becchio A, Giannarini L, Maggi E, Pupilli C, Tonelli F, Romagnani S. Type 1 T-helper cell predominance and interleukin-12 expression in the gut of patients with Crohn's disease. Am J. Pathol., 1997; 150(3):823-32.
    • Plevy S E, Landers C J, Prehn J, Carramanzana N M, Deem R L, Shealy D, Targan S R. A role for TNF-alpha and mucosal T helper-1 cytokines in the pathogenesis of Crohn's disease. J. Immunol., 1997; 159(12):6276-82.
    • Simpson S J, Shah S, Comiskey M, de Jong Y P, Wang B, Mizoguchi E, Bhan A K, Terhorst C. T cell-mediated pathology in two models of experimental colitis depends predominantly on the interleukin 12/Signal transducer and activator of transcription (Stat)-4 pathway, but is not conditional on interferon gamma expression by T cells. J Exp Med., 1998; 187(8):1225-34.
    • Trinchieri G. Interleukin-12: a cytokine at the interface of inflammation and immunity. Adv Immunol, 1998; 70:83-243.
    • Tuschl T, Zamore P D, Lehmann R, Bartel D P, Sharp P A. Targeted mRNA degradation by double-stranded RNA in vitro. Genes Dev., 1999; 13(24):3191-7.
    • Vandenbroeck K, Alloza I, Gadina M, Matthys P. Inhibiting cytokines of the interleukin-12 family: recent advances and novel challenges. J Pharm Pharmacol., 2004, 56(2):145-60.
    • Wianny F, Zernicka-Goetz M. Specific interference with gene function by double-stranded RNA in early mouse development. Nat Cell Biol., 2000, 2(2):70-5.
    • Williams B R. Role of the double-stranded RNA-activated protein kinase (PKR) in cell regulation. Biochem Soc Trans, 1997, 25(2):509-13.

Claims (27)

1-47. (canceled)
48. A method of treating an intestinal disorder in an individual, comprising administering to a patient a therapeutically effective amount of a short interfering nucleic acid molecule (siNA), wherein the siNA molecule targets Interleukin 12.
49. The method according to claim 48, wherein the siNA molecule is administered intrarectally.
50. The method of claim 48, wherein the disorder is localized in an organ selected from the group consisting of the small intestine, the large intestine, and the rectum.
51. The method of claim 48, wherein the disorder is a disorder of the small intestine.
52. The method of claim 48, wherein the disorder is selected from the group consisting of hyperproliferative diseases, autoimmune and inflammatory bowel diseases (IBD), infectious diseases of the intestine, malabsorption syndromes, rectal diseases and diarrhea.
53. The method of claim 48, wherein the disorder is selected from the group consisting of colorectal cancer, Crohn's disease, colitis, ulcerative colitis, irritable bowel syndrome, pseudomembranous colitis, amebiasis, intestinal tuberculosis, colonic polyps, diverticular disease, constipation, and intestinal obstruction.
54. The method of claim 48, wherein the siNA molecule is selected from the group consisting of dsRNA, siRNA, and shRNA.
55. The method of claim 48, wherein the siNA molecule is dsRNA.
56. The method of claim 48, wherein the siNA molecule modulates miRNA levels.
57. The method of claim 48, wherein the siNA molecule comprises a modified oligonucleotide.
58. The method of claim 48, wherein the siNA molecule is 40 base pairs or fewer in length.
59. The method of claim 48, wherein the siNA molecule has 3′ overhangs.
60. The method of claim 59, wherein the 3′ overhangs are dinucleotides.
61. The method of claim 60, wherein the dinucleotide overhangs are made of thymidine nucleotides.
62. The method of claim 48, wherein a plurality of siNA molecules is used, and wherein the siNA molecules are targeted to the same mRNA sequences or to different mRNA sequences.
63. The method of claim 48, wherein the siNA molecule comprises a sequence selected from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 85.
64. The method of claim 48, wherein the siNA molecule is targeted to the Interleukin-12 35 kDa subunit (IL12-p35).
65. The method of claim 64, wherein the siNA molecule comprises a nucleotide sequence selected from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 33.
66. The method of claim 48, wherein the siNA molecule is targeted to the Interleukin-12 40 kDa subunit (IL12-p40).
67. The method of claim 66, wherein the siNA molecule comprises a nucleotide sequence selected from the group consisting of SEQ ID NO: 34 to SEQ ID NO: 85.
68. A method of treating an intestinal disorder in an individual, comprising administering intrarectally to a patient a therapeutically effective amount of a short interfering nucleic acid molecule (siNA) that targets a gene associated with said intestinal disorder.
69. The method of claim 68, wherein the disorder is selected from the group consisting of hyperproliferative diseases, autoimmune and inflammatory bowel diseases (IBD), infectious diseases of the intestine, malabsorption syndromes, rectal diseases and diarrhea.
70. The method of claim 68, wherein the disorder is selected from the group consisting of colorectal cancer, Crohn's disease, colitis, ulcerative colitis, irritable bowel syndrome, pseudomembranous colitis, amebiasis, intestinal tuberculosis, colonic polyps, diverticular disease, constipation, and intestinal obstruction.
71. A pharmaceutical composition for the treatment of an intestinal disorder comprising a short interfering nucleic acid molecule (siNA) compound formulated for intrarectal administration.
72. The pharmaceutical composition of claim 71, wherein the siNA molecule targets IL-12 mRNA expression.
73. The pharmaceutical composition of claim 72, wherein the siNA molecule comprises a sequence selected from SEQ ID NO: 1 to SEQ ID NO: 85.
US11/908,696 2005-03-14 2006-03-14 Treatment of Intestinal Conditions Abandoned US20110313016A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GBGB0505081.0A GB0505081D0 (en) 2005-03-14 2005-03-14 Downregulation of interleukin-12 expression by means of rnai technology
GB0505081.0 2005-03-14
PCT/GB2006/050051 WO2006097768A2 (en) 2005-03-14 2006-03-14 Treatment of intestinal conditions by a compound which causes rna interference

Publications (1)

Publication Number Publication Date
US20110313016A1 true US20110313016A1 (en) 2011-12-22

Family

ID=34508943

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/908,696 Abandoned US20110313016A1 (en) 2005-03-14 2006-03-14 Treatment of Intestinal Conditions
US11/858,579 Active 2027-01-28 US7807650B2 (en) 2005-03-14 2007-09-20 Methods and compositions for the treatment of intestinal conditions
US12/871,366 Active US8067577B2 (en) 2005-03-14 2010-08-30 Methods and compositions for the treatment of intestinal conditions

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/858,579 Active 2027-01-28 US7807650B2 (en) 2005-03-14 2007-09-20 Methods and compositions for the treatment of intestinal conditions
US12/871,366 Active US8067577B2 (en) 2005-03-14 2010-08-30 Methods and compositions for the treatment of intestinal conditions

Country Status (12)

Country Link
US (3) US20110313016A1 (en)
EP (2) EP2386298B1 (en)
JP (2) JP2008532540A (en)
CN (2) CN103432592B (en)
AU (1) AU2006224338B2 (en)
BR (1) BRPI0609323A2 (en)
CA (1) CA2599220C (en)
ES (1) ES2587252T3 (en)
GB (1) GB0505081D0 (en)
MX (1) MX2007011235A (en)
RU (1) RU2418594C2 (en)
WO (1) WO2006097768A2 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2008232316A1 (en) * 2007-03-26 2008-10-02 Newcastle Innovation Limited Therapeutic targets and molecules
WO2009088849A2 (en) * 2008-01-04 2009-07-16 Immune Disease Institute, Inc. Treatment or prevention of inflammation by targeting cyclin d1
US20130274317A1 (en) 2010-11-04 2013-10-17 Sandra Milena Ocampo Derivatives of small interfering rnas and use thereof
US9248120B2 (en) 2011-08-23 2016-02-02 The Board Of Trustees Of The Leland Stanford Junior University Reversing intestinal inflammation by inhibiting retinoic acid metabolism
US10499908B2 (en) 2014-03-04 2019-12-10 Maquet Cardiovascular Llc Surgical implant and method and instrument for installing the same
US20150250474A1 (en) 2014-03-04 2015-09-10 Maquet Cardiovascular Llc Surgical implant and method and instrument for installing the same
CN109414414A (en) 2016-03-16 2019-03-01 戴维·格拉德斯通研究所 Method and composition for treating obesity and/or diabetes and for identifying candidate therapeutic agent
RU2676652C1 (en) * 2018-02-28 2019-01-09 Федеральное государственное бюджетное учреждение "Детский научно-клинический центр инфекционных болезней Федерального медико-биологического агентства" Method of treatment of intestinal infections in children

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040029275A1 (en) * 2002-08-10 2004-02-12 David Brown Methods and compositions for reducing target gene expression using cocktails of siRNAs or constructs expressing siRNAs

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1469009A2 (en) * 1998-05-21 2004-10-20 Isis Parmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
US6448081B1 (en) * 2001-05-07 2002-09-10 Isis Pharmaceuticals, Inc. Antisense modulation of interleukin 12 p40 subunit expression
IL151928A0 (en) * 2000-03-30 2003-04-10 Whitehead Biomedical Inst Rna sequence-specific mediators of rna interference
JP4095895B2 (en) * 2000-12-01 2008-06-04 マックス−プランク−ゲゼルシャフト ツール フォーデルング デル ヴィッセンシャフテン エー.ヴェー. Short RNA molecules that mediate RNA interference
US20050148530A1 (en) 2002-02-20 2005-07-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
AU2003207708A1 (en) 2002-02-20 2003-09-09 Sirna Therapeutics, Inc. Rna interference mediated inhibition of map kinase genes
GB2406568B (en) 2002-02-20 2005-09-28 Sirna Therapeutics Inc RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US20040115641A1 (en) 2002-12-11 2004-06-17 Isis Pharmaceuticals Inc. Modulation of ROCK 1 expression
CA2500224C (en) 2002-09-25 2015-04-28 University Of Massachusetts In vivo gene silencing by chemically modified and stable sirna
KR20050083855A (en) 2002-11-01 2005-08-26 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Compositions and methods for sirna inhibition of hif-1 alpha
EP2314691A3 (en) * 2002-11-14 2012-01-18 Dharmacon, Inc. Fuctional and hyperfunctional siRNA
EP1631669A2 (en) * 2003-04-09 2006-03-08 Biodelivery Sciences International, Inc. Cochleate compositions directed against expression of proteins
US20050013854A1 (en) * 2003-04-09 2005-01-20 Mannino Raphael J. Novel encochleation methods, cochleates and methods of use
WO2005044976A2 (en) 2003-06-20 2005-05-19 Isis Pharmaceuticals, Inc. Oligomeric compounds for use in gene modulation
WO2005045037A2 (en) 2003-10-23 2005-05-19 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF 5-ALPHA REDUCTASE AND ANDROGEN RECEPTOR GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
SE0303397D0 (en) * 2003-12-17 2003-12-17 Index Pharmaceuticals Ab Compounds and method for RNA interference
US20050272682A1 (en) * 2004-03-22 2005-12-08 Evers Bernard M SiRNA targeting PI3K signal transduction pathway and siRNA-based therapy
TW200639253A (en) 2005-02-01 2006-11-16 Alcon Inc RNAi-mediated inhibition of ocular targets

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040029275A1 (en) * 2002-08-10 2004-02-12 David Brown Methods and compositions for reducing target gene expression using cocktails of siRNAs or constructs expressing siRNAs

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
http://www.merriam-webster.com/dictionary/intestine 01/22/2013. *
Lawrence et al. (Gastroenterology, 2003 Vol. 125, Issue 6: 1750-1761). *
Schmidt et al. (Pathobiology 2002, Vol. 70(3): 177-183). *

Also Published As

Publication number Publication date
CN103432592B (en) 2017-05-17
US20100317720A1 (en) 2010-12-16
CA2599220C (en) 2018-05-22
EP1868592A2 (en) 2007-12-26
RU2418594C2 (en) 2011-05-20
WO2006097768A2 (en) 2006-09-21
GB0505081D0 (en) 2005-04-20
CA2599220A1 (en) 2006-09-21
EP2386298B1 (en) 2016-07-06
JP2008532540A (en) 2008-08-21
RU2007138031A (en) 2009-04-20
EP2386298A1 (en) 2011-11-16
AU2006224338B2 (en) 2011-01-06
US8067577B2 (en) 2011-11-29
WO2006097768A3 (en) 2007-03-29
CN103432592A (en) 2013-12-11
MX2007011235A (en) 2007-11-14
AU2006224338A1 (en) 2006-09-21
CN101184480A (en) 2008-05-21
US20080249048A1 (en) 2008-10-09
BRPI0609323A2 (en) 2010-03-16
JP2012255016A (en) 2012-12-27
JP5756441B2 (en) 2015-07-29
US7807650B2 (en) 2010-10-05
ES2587252T3 (en) 2016-10-21

Similar Documents

Publication Publication Date Title
JP5756441B2 (en) Treatment of bowel disease
US8871729B2 (en) Treatment of CNS conditions
EP3210611B1 (en) Methods of treating vascular inflammatory disorders
RU2718534C2 (en) Mir-155 inhibitors for treatment of cutaneous t-cell lymphoma (ctcl)
AU2017276806A1 (en) Methods of treating neuroblastoma and reagents therefor
RU2410430C2 (en) Methods and compositions for inhibiting expression of p2x7 receptor
WO2019207066A1 (en) Methods and compositions for the treatment of sjögren's syndrome
WO2024051765A1 (en) T cell with silenced cd59 gene and use thereof
WO2019023344A1 (en) Methods and compositions for treating and preventing metastatic tumors
US8889648B2 (en) Nucleic acid having an anti-metabolic syndrome effect
US20230374505A1 (en) Human XIST Antisense Oligonucleotides for X Reactivation Therapy

Legal Events

Date Code Title Description
AS Assignment

Owner name: SYLENTIS S.A.U., SPAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JIMENEZ, ANA I.;GASCON, IRENE;JIMENEZ, MARIA CONCEPCION;AND OTHERS;SIGNING DATES FROM 20071005 TO 20071023;REEL/FRAME:020029/0430

STCB Information on status: application discontinuation

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