WO2012094115A1 - Compositions and methods for inhibiting expression of flt3 genes - Google Patents

Compositions and methods for inhibiting expression of flt3 genes Download PDF

Info

Publication number
WO2012094115A1
WO2012094115A1 PCT/US2011/064896 US2011064896W WO2012094115A1 WO 2012094115 A1 WO2012094115 A1 WO 2012094115A1 US 2011064896 W US2011064896 W US 2011064896W WO 2012094115 A1 WO2012094115 A1 WO 2012094115A1
Authority
WO
WIPO (PCT)
Prior art keywords
acid molecule
double
nucleotide
ribonucleic acid
stranded ribonucleic
Prior art date
Application number
PCT/US2011/064896
Other languages
French (fr)
Inventor
John Frederick Boylan
Birgit Bramlage
Wei He
Markus Hossbach
Original Assignee
Arrowhead Research Corporation
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 Arrowhead Research Corporation filed Critical Arrowhead Research Corporation
Publication of WO2012094115A1 publication Critical patent/WO2012094115A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.

Definitions

  • This invention relates to double-stranded ribonucleic acids (dsRNAs), and their use in mediating RKA Interference to Inhibit the expression of the FLT3 gene. Furthermore, the use of said dsRNA to treat inflammation and proliferative disorders, like cancers, is part of this invention.
  • Cancer remains an important area of high unmet medical need. The majority of current treatments provide small gains in overall survival requiring a delicate balance between efficacy and toxicity. Cancer is characterized by uncontrolled growth and survival driven by the improper regulation of proliferation.
  • the frns ⁇ Hke receptor tyrosine kinase-3 (FLT3) plays a key role in the differentiation and growth of immature hematopoietic stem cells and immune cells.
  • FLT3 frns ⁇ Hke receptor tyrosine kinase-3
  • Activating mutations in the FLT3 gene create a protein that is constitulively active relaying a constant pro-proSiferative signal in mutant leukemia cells. The presence of
  • a specific FLT3 siRNA is expected to reduce the level of FLT3 mRNA leading to a loss in protein and elimination of STATS phosphorylation. This will produced a loss of growth and an increase in cell death in FLT3 mutant expressing leukemia cells and a reversible growth arrest in wild-type expressing ceils,
  • the invention provides double-stranded ribonucleic acid molecules (dsRJMAs), as well as compositions and methods for inhibiting the expression of the FLT3 gene, in particular the expression of the FLT3 gene, in a ceil tissue or mammal using such dsRNA.
  • the invention also provides compositions and methods for treating pathological conditions and diseases caused by the expression of the FLT3 gene such as in proliferative disorders like cancer and inflammation.
  • the invention provides double-stranded ribonucleic acid (dsRNA) molecules able to selectively and efficiently decrease the expression of FLT3,
  • dsRNAi provides a method for the therapeutic and/or prophylactic treatment of diseases/disorders which are associated with inflammation and proliferative disorders, like cancers.
  • Particular disease/disorder states include the therapeutic and/or prophylactic treatment of inflammation, like fibrosis, and proliferative disorders, like cancers, which method comprises administration of dsRNA targeting FLT3 to a human being or animal in one preferred embodiment the described dsRNA molecule is capable of inhibiting the expression of a FLT3 gene by at least 60%, preferably by at least 70 %, most preferably by at least 80 %.
  • the invention provides double-stranded ribonucleic acid (dsRNA) molecules for inhibiting the expression of a FLT3 gene, in particular the expression of the mammalian or human FLT3 gene.
  • the dsRNA comprises at least two sequences that are complementary to each other.
  • the dsRNA comprises a sense strand comprising a first sequence and an antisense strand comprising a second sequence, see sequences provided in the sequence listing and also the specific dsRNA pairs in the appended table 1 and table 2.
  • the sense strand comprises a sequence which has an identity of at least 90% to at least a portion of an mRNA encoding FLT3, Said sequence is located in a region of complementarity of the sense strand to the antisense strand, preferably within n cleotides 2-7 of the 5* terminus of the antisense strand, In one preferred embodiment the dsRNA specifically targets the human FLT3 gene.
  • the antisense strand comprises a nucleotide sequence which is substantially complementary to at least part of an mRNA encoding said FLT3 gene, and the region of complementarity is most preferably less than 30 nucleotides in length.
  • the length of the herein described Inventive dsRNA molecules is in the range of about 16 to 30 nucleotides, in partictdar in the range of about 18 to 28 nucleotides.
  • Particularly useful in context of this invention are duplex lengths of about 19, 20. 21, 22, 23 or 24 nucleotides. Most preferred are duplex stretches of 19, 21 or 23 nucleotides.
  • the dsR A upon delivery to a cell expressing a FLT3 gene, inhibits the expression of a FLT3 gene in vitro by at least 60%, preferably by at least 70%, and most preferably by 80%
  • Appended Table 1 relates to preferred molecules to be used as dsRNA in accordance with this invention.
  • modified dsRNA molecules are provided herein and are in particular disclosed in appended table 2, providing illustrative examples of modified dsRNA molecules of the present invention.
  • Table 2 provides for illustrative examples of modified dsRNAs of this invention (whereby the corresponding sense strand and antisense strand is provided in this table).
  • Table 5 The relation of the unmodified preferred molecules shown in Table 1 to the modified dsRNAs of Table 2 is illustrated in Table 5.
  • the illustrative modifications of these constituents of the inventive dsRNAs are provided herein as examples of modifications.
  • Tables 3 and 4 provide for selective biological, clinical and pharmaceutical relevant parameters of certain dsRNA molecules of this invention.
  • the preferred dsRNA molecules are provided in the appended table 1 and, inter alia and preferably, wherein the sense strand is selected from the group consisting of the nucleic acid sequences depicted in SEQ ID NOs: 3, 5, 7, 1 1, 13, 21, 25 and 2? and the antisense strand is selected from the group consisting of the nucleic acid sequences depleted in SEQ ID NOs: 4, 6, 8, 12, 14, 22, 26 and 28 Accordingly, the inventive dsRNA molecule may, Inter alia, comprise the sequence pairs selected from the group consisting of SEQ ID NOs: 3/ 4 5 5/6, 7/8, 1 1 12, 13/14, 25/22, 25/26 and 27/28.
  • pairs of $F,Q D NOs relate to corresponding sense and antisense strands sequences (5' to 3') as also shown in the tables.
  • the dsRNA molecules comprise an antisense strand with a T overhang of 1-5 nucleotides In length, preferably 1 -2 nucleotides in length.
  • said overhang of the antisense strand comprises uracil or nucleotides which are complementary to the mRNA encoding FLT.I. !
  • said dsRNA molecules comprise a sense strand with a 3* overhang of 1-3 nucleotides in length, preferably 1-2 nucleotides in length.
  • said overhang of the sense strand comprises uracil or nucleotides which are identical to the mRNA encoding FLT3.
  • the dsRNA molecules comprise a sense strand with a 3' overhang of 1-5 nucleotides in length, preferably 1-2 nucleotides in length, and an antisense strand with a 3 * overhang of 1-5 nucleotides in length, preferably 1-2 nucleotides in length.
  • said overhang of the sense strand comprises uracil or nucleotides which are at least 90% identical ⁇ the nsRNA encoding FLT3 and said overhang of the antisense strand comprises uracil or nucleotides which are at least 90% complementary to the RNA encoding FLT3.
  • the ds A molecules of the invention may be comprised of naturally occurring nucleotides or may be comprised of at least one modified nucleotide, such as a 2" ⁇ 0 ⁇ methyi 5 modified nucleotide, inverted deox thymidine, a nucleotide comprising a 5 ! ⁇ phosphorothioa ⁇ e group, and a terminal nucleotide linked to a choiesteryl derivative or dodecanoic acid blsdec lamide group.
  • 2' modified nucleotides may have the additional advantage that certain immunosiimulatory factors or cytokines are suppressed when the inventive dsRNA molecules are employed in vivo, for example in a medical setting, Alternatively and non-limiting, the
  • modified nucleotide may be chosen from the group of: a 2' ⁇ deoxy-2 1 -fltioro modified nucleotide, a 2 ! -deoxy ⁇ modified nucleotide, a locked nucleotide, an abasie nucleotide, 2' ⁇ arnino ⁇ modified nucleotide, 2 5 -a!kyl ⁇ modified nucleotide, morpholino nucleotide, a phosphoramidate, and a non- natural base comprising nucleotide.
  • the dsRNA molecules comprises at least one of the following modified nucleotides: a 2' ⁇ Omethyl modified nucleotide,
  • I S a nucleotide comprising a 5' «phosphoiothioate group and a deoxythymidsne.
  • Preferred dsRNA molecules comprising modified nucleotides are given in table 2.
  • one of those deoxythyrmdme nucleotides at the 3' of both strand is a inverted deoxythymi ine,
  • inventive dsRNA molecules comprise modified 0 nucleotides as detailed in the sequences given in table 2.
  • inventive dsRNA molecule comprises sequence pairs selected from the group consisting of SEQ ID NOs: 3/ 4, 5/6, 7/8, 1 1/12, 13/14, 21/22, 25/26 and 27/28, and comprises overhangs at the antisense and'' or sersse strand of i ⁇ 2 deoxythymidines,
  • inventive dsRNA molecule comprises sequence pairs selected from the group consisting of SEQ 5 ID NOs: 3/ 4, 5/6, 7/8, 1 1/12, 13/14, 21/22, 25/26 and 27/28, and comprise modifications as detailed in table 2.
  • Preferred dsRNA molecules comprising modified nucleotides are listed in table 2-4, with the most preferred dsRNA molecules depicted in SEQ ID Nos: 149/150, 151/152, 153/154, 157/158, 159/160, 567/168, 171/172, 173/174, 293/294, 295/29 nd 297/298.
  • the inventive dsRNAs comprise modified nucleotides on 0 positions different from those disclosed in table 2.
  • two deoxythymidine nucleotides are found at the 3 * of both strands of the dsRNA molecule, Preferably said deoxythymidine nucleotides form ais overhang.
  • the dsRNA molecules of the Invention comprise a sense and an antisense strand wherein both strands have a half-Hie of at least 0.9 hoars, Irs one preferred embodiment the dsRNA molecules of the invention comprise a sense and an antisense strand wherein both strands have a half-life of at least 48 hours, preferably in human serum.
  • the dsRNA molecules of the invention are non-lmmunestimulatory, e.g. do not stimulate IMF-alpha and TNF-alpha in vitro. In another embodiment the dsRNA molecules of the invention do stimulate INF-alpha and TNF-alpha in vitro to a very minor degree.
  • a nucleic acid sequence encoding a sense strand and / or an antisense strand comprised in the dsRNAs as defined herein are provided.
  • the invention also provides tor ceils comprising at least one of the dsRNAs of the invention.
  • the cell is preferably a mammalian cell, such as a human cell.
  • tissues and/or non-human organisms comprising the herein defined dsRNA molecules are an embodiment of this invention, whereby said non-human organisms are particularly useful for research purposes or as research tools, for example in drug testing.
  • the invention relates to a method for inhibiting the expression of a FLT3 gene, in particular a mammalian or human FLT3 gene, In a cell, tissue or organism comprising the following steps:
  • step (a) introducing into the DCi > tissue or organism a double-stranded ribonucleic acid (dsRNA) as defined herein; and (b) maintaining said cell, tissue or organism produced In step (a) for a time sufficient to obtain degradation of the mRNA transcript of a FLT3 gene, thereby inhibiting expression of a FLT3 gene in a given cell.
  • dsRNA double-stranded ribonucleic acid
  • the invention also relates to pharmaceutical compositions comprising the inventive dsRNAs of the invention. These pharmaceutical compositions are particularly usefui in the Inhibition of the expression of a FLT3 gene In a vash, a tissue or an organism.
  • the pharmaceutical composition comprising one or more of the dsRNA of the invention may also comprise (a) pharmaceutically acceptable earrier(s), diiuent(s) and/or excipient(s),
  • the invention provides methods for treating, preventing or managing inflammation and / or proliferative disorders like cancers which are associated with FLT3, said method comprising administering to a subject in need of such treatment, prevention or management a therapeutically or prophylactiealiy effective amount of ne or more of the dsRNAs of the invention.
  • said subject is a mammal, most preferably human patient.
  • the invention provides a method for treating a subject having a pathological condition mediated by the expression of a FLT3 gene.
  • Such conditions comprise disorders associated with inflammation and. proliferative disorders, like cancers, as described above.
  • the dsRNA acts as a therapeutic agent for controlling the expression of a FLT3 gene
  • the method comprises administering a pharmaceutical composition of the invention to the patient (e.g., human), such that expression of a FLT3 gene is silenced.
  • the dsRNAs of the invention specifically target mRNAs of a FLT3 gene.
  • the described dsRNAs specifically decrease FLT3 niRMA levels and do not directly affect the expression and / or m NA levels of off-target genes in the ceil.
  • the described dsRNA decrease FLT3 mRNA levels in the liver by at least 60%, preferably by at least 70%, most preferably by at least 80% in vivo. In another embodiment the described dsRNAs decrease FLT3 mRNA levels in vivo for at least 4 days. In another preferred embodiment, the dsRNAs of the invention are used for the preparation of a pharmaceutical composition for the treatment of inflammation and proliferative disorders, like cancer. Cancers to be treated with said pharmaceutical composition comprise but are not limited to leukemia and myeloproliferative diseases.
  • the invention provides vectors for inhibiting the expression of a FLT3 gene in a cell, in particular a FLT3 gene comprising a regulatory sequence operahly linked to a nucleotide sequence that encodes at least one sti'and of the dsRNA molecules of the invention.
  • the invention provides a cell comprising a vector for inhibiting the expression of a FLT3 gene in a cell.
  • Said vector comprises a regulatory sequence operahly linked to a nucleotide sequence that encodes at least one strand of the dsRNA molecule of the invention, Yet, it is preferred that said vector comprises, besides said regulatory sequence a sequence that encodes at least one "sense strand" of the inventive dsRNA and at least one "anti sense strand” of said dsRNA.
  • the claimed cell comprises two or more vectors comprising, besides said regulator ⁇ -' sequences, the herein defined seqneoce(s) that encode(s) at least one strand of the dsRNA molecule of the invention.
  • the method comprises administering a composition comprising a dsRMA, wherein the dsKNA comprises a nucleotide sequence which is complementary to at least a part of an RNA transcript of a FLT3 gene of the mammal to be treated.
  • dsKNA comprises a nucleotide sequence which is complementary to at least a part of an RNA transcript of a FLT3 gene of the mammal to be treated.
  • vectors and cells comprising nucleic acid molecules that encode for at least one strand of the herein defined dsRNA molecules can be used as pharmaceutical compositions and may, therefore, also be employed in the herein disclosed methods of treating a subject in need of medical intervention, it is also of note that these embodiments relating to pharmaceutical compositions and to corresponding methods of treating a (human) subject also relate to approaches like gene therapy approaches.
  • FLT3 specific dsRNA molecules as provided herein or nucleic acid molecules encoding individual strands of these inventive dsRNA molecules may also be inserted into vectors and used as gene therapy vectors for human patients.
  • Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Patent 5,328,470) or by stereotactic injection (see e.g., Chen et al (1994) Proc. Natl Acad. ScL USA 91 :3054-3057) addict
  • the pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the complete gene deliver)' vector can be produced intact from recombinant ceils, e.g.. retroviral vectors, the pharmaceutical preparation can include or»e or more ceils which produce the gene delivery system.
  • FLT3 specific dsRNA molecules thai modulate FLT3 gene expression activity are expressed from transcription units inserted into DNA or R A vectors (see, e.g. f Skiicht, A,, et al. International PCX Publication No. WO 00/221 13).
  • These transgenes can be Introduced as a linear construct. & circular piasmid. or a viral vector, which can be Incorporated and inherited as a iransgene integrated into the host genome.
  • the transgene can also be constructed to permit it to be inherited as an extraehromosomal piasmid (Gassmann, et ai, Proc. Natl. Acad. Sci, USA ( 1 95) 92: i 292).
  • a dsRNA can be transcribed by promoters on two separate expression vectors and co-transfected into a target cell.
  • each individual strand of the dsRNA can be transcribed by promoters both of which are located on the same expression piasmid.
  • a dsRNA is expressed as an inveited repeat joined by a linker polynucleotide sequence such that the dsRNA has a stem and loop structure.
  • the recombinant dsRNA expression vectors are preferably DNA p! asm ids or viral vectors.
  • dsRNA expressing viral vectors can be constructed based on, but not limited to, adeno- associated vims (for a review, see Muzyezka, et al., Curr. Topics Micro. Immunol (1 92) 158:97-129)); adenovirus (see, for example, Berkner, et al. s BioTechmqtses (1998) 6:616), Rosenfeld et al, (1991 , Science 252:431 -434), and Rosenfeld et al. (1992), Cell 68: 143-155)); or alphavirus as well as others known in the art.
  • Retroviruses have beers used to introduce a variety of genes into many different cell, types, including epithelial cells, in vitro and/or in vivo (see, e.g., Danes and Mulligan, Proc. Natl Acad. Sci. USA (1998) 85:6460-6464), Recombinant retroviral vectors capable of transducing and expressing genes inserted into the genome of a ceil can be produced by transacting the recombinant retroviral genome into suitable packaging cell lines such as PA317 and Psi ⁇ CRlP (Comette et al., 1991, Human Gene Therapy 2:5-10; Cone et aL, 1 84, Proc, Natl Acad. Sci. USA 81 :6349).
  • suitable packaging cell lines such as PA317 and Psi ⁇ CRlP (Comette et al., 1991, Human Gene Therapy 2:5-10; Cone et aL, 1 84, Proc, Natl Acad. Sci. USA
  • Recombinant adenoviral vectors can be used to infect a wide variety of cells and tissues in susceptible hosts (e.g., rat, hamster, dog, and chimpanzee) (Hsu et al, 1992, J. Infectious Disease, 166:769), and also have the advaniage of not requiring mitoticalSy active cells for infection.
  • susceptible hosts e.g., rat, hamster, dog, and chimpanzee
  • the promoter driving dsR A expression in either a DNA plasrnid or viral vector of the invention may be a eukaryotic R A polymerase 1 (e.g. ribosomal RNA promoter), RNA polymerase 11 (e.g. CMV early promoter or actio promoter or Ul snRNA promoter) or preferably RNA polymerase Hi promoter (e.g. U6 snRNA or 7S RNA promoter) or a prokaryotic promoter, for example the T7 promoter, provided the expression plasrnid also encodes T7 RNA polymerase required for transcription from a T7 promoter.
  • the promoter can also direct transgene expression to the pancreas (see, e.g. the insulin regulatory sequence for pancreas (Bucchini et al, 1 86, Proc. Nail Acad. Sci. USA 83:251 ⁇ -2515)).
  • expression of the transgene can be precisely regulated, for example, by using an inducible regulatory sequence and expression systems such as a regulatory sequence that is sensitiv to certain physiological regulators, e.g., circulating glucose levels, or hormones (Doeherty et al, 1994, FASEB J. 8:20-24).
  • inducible expression systems suitable for the control of transgene expression in cells or in mammals include regulation by eedysone, by estrogen, progesterone, tetracycline, chemical inducers of dimerhaiion, and isopropyl-bela-Dl - thiogalaetopyranos ide (EPTG).
  • recombinant vectors capable of expressing dsRMA molecules are delivered as described below, and persist in target cells.
  • viral vectors can be used that provide for transient expression of dsRNA molecules. Such vectors can be repeatedly administered as necessary.
  • dsRNA expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex-planted from the patient followed by emtroduction into the patient, or by any other means that allows for introduction into a desired target cell dsRNA expression
  • DNA plasm ids are typically traosfected into target cells as a complex with cation 3C lipid carriers (e.g. Gligofeetamme) or non-eationic lipid-based carriers (e,g, Transk ⁇ TKO m ).
  • lipid transfections for dsRNA-mediated knockdowns targeting different regions of a single FLT3 gene or multiple FLT3 genes over a period of a week or more are also contemplated by the invention.
  • Successful introduction of the vectors of the invention into host cells can be monitored using various known methods.
  • transient transfection can be signaled with a reporter, such as a fluorescent marker, such as Green Fluorescent Protein (GFP), Stable transfection of ex vivo cells can be ensured using markers that provide the transfected ceil with resistance to specific environmental factors (e.g., antibiotics and drugs), such as hygromycin B resistance.
  • a reporter such as a fluorescent marker, such as Green Fluorescent Protein (GFP)
  • Stable transfection of ex vivo cells can be ensured using markers that provide the transfected ceil with resistance to specific environmental factors (e.g., antibiotics and drugs), such as hygromycin B resistance.
  • the following detailed description discloses how to make and use the dsRNA and compositions containing dsRNA to inhibit the expression of a target FLT3 gene, as well as compositions and methods for treating diseases and disorders caused by the expression of said FLT3 gene.
  • G,” “C,” “A”, “U” and “T” or “dT” respectively each generally stand for a nucleotide that contains guanine, cytosine, adenine, uracil and deoxythymidine as a base, respectively.
  • ribonucleotide or “nucleotide” can also refer to a modified nucleotide, as further detailed below, or a surrogate replacement moiety. Sequences comprising such replacement moieties are embodiments of the invention.
  • the herein described dsRNA molecules may also comprise "overhangs", i.e.
  • unpaired, overhanging nucleotides which are ot directly involved in the UNA double helical structure normally formed by the herein deimed pair of "sense strand” and "anti sense strand", Often, such an overhanging stretch comprises the deoxythymidine nucleotide, in most embodiments, 2 deosyihymidines in the 3' end, Such overhangs will be described and illustrated below.
  • FLT3 as used herein relates in particular to the frns-like receptor tyrosine kinase 3 said term relates to the corresponding gene, encoded mRNA, encoded prolem/poiypeptide as well as functional fragments of the same, Preferred is the human FLT3 gene.
  • the ds NAs of th invention target the FLT3 gene of human (H.sapiens) and eynornoigous monkey (Macaca iascicularis) FLT3 gene.
  • dsRMAs targeting the rat (Rattus norvegicus) and mouse (Miss musculus) FLT3 gene are part, of this invention.
  • FLT3 gene/sequence does not only relate to (the) wild-type sequence(s) hut also to mutations and alterations which may be comprised in said gene/sequence. Accordingly, the present invention is not limited to the specific dsRNA molecules provided herein, The invention also reiaies to dsRNA molecules that comprise an antisense strand that is at least 85% complementary to the corresponding nucleotide stretch of a RNA transcript of a FLT3 gene that comprises such mu lons aJterations,
  • target sequence refers to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during the transcription of a FLT3 gene, including mRNA that is a product of RNA processing of a primary transcription product.
  • strand comprising a sequence refers to an oligonucleotide comprising a chain of nucleotides that is described by the sequence referred to using the standard nucleotide nomenclature. However, as detailed herein, such a “strand comprising a sequence” may also comprise modifications, like modified nucleotides.
  • complementary when used to describe a first nucleotide sequence in relation to a second nucleotide sequence, refers to the ability of an oligonucleotide or polynucleotide comprising the first nucleotide sequence to hybridize and form a duplex structure under certain conditions with an oligonucleotide or polynucleotide comprising the second nucleotide sequence.
  • “Complementary” sequences, as used herein may also include, or be formed entirely from, non- Watson-Crick base pairs and/or base pairs formed from non-natural and modified nucleotides, in as tar as the above requirements with respect to their ability to hybridize are fulfilled,
  • Sequences referred to as "fully complementary” comprise base-pairing of the oligonucleotide or polynucleotide comprising the first nucleotide sequence to the oligonucleotide or polynucleotide comprising the second nucleotide sequence over the entire length of the first and second nucleotide sequence.
  • first sequence is referred to as “substantially complementary” with respect to a second sequence herein
  • the two sequences can be Miy complementary, or they may form one or more, but preferably not more than 13 mismatched base pairs upon hybridization
  • the terms “complementary”, “fully complementary” and “substantially complementary” herein may be used with respect to the base matching between the sense strand and the antisense strand of a dsR A, or between the andsense strand of a dsRNA and a target sequence, as will be understood from the context of their use.
  • double-stranded NA M refers to a ribonucleic acid molecule, or complex of ribonucleic acid molecules, having a duplex structure comprising two anti-parallel and substantially complementary nucleic acid strands.
  • the iwo strands forming the duple structure may he different portions of one larger RNA molecule, or they may be separate RNA molecules. Where the two strands are part of one larger molecule, and therefore are connected by an uninterrupted chain of nucleotides between the 3 '-end of one strand and the 5' end of the respective other strand forming the duplex structure, the connecting RNA chain is referred to as a "hairpin loop".
  • RNA strands may have the same or a different number of nucleotides.
  • a dsRNA may comprise one or more nucleotide overhangs.
  • the nucleotides in said "overhangs” may comprise between 0 and 5 nucleotides, whereby “0 s * means no additional nucieotide(s) that form(s) an "overhang” and whereas ;i 5" means ftve additional nucleotides on the individual strands of the dsRNA duplex. These optional "overhangs” are located in the 3 5 end of the individual strands. As will be detailed below, also dsRNA molecules which comprise only an "overhang” in one of the two strands may be useful and even advantageous in context of this invention, The "overhang” comprises preferably between 0 and 2 nucleotides.
  • nucleotide overhang refers to the unpaired nucleotide or nucleotides that protrude from the duplex structure of a dsRNA when a 3' ⁇ end of one strand of the dsRNA extends beyond the 5'-end of the other strand, or vice versa.
  • the antisense strand comprises 23 nucleotides and the sense strand comprises 21 nucleotides, forming a 2 nucleotide overhang at the 3' end of the antisense strand.
  • the 2 nucleotide overhang is fully complementary to the m NA of the target gene.
  • “Blunt” or “blunt end” means that there are no unpaired nucleotides at that end of the dsRNA, i.e., no nucleotide overhang
  • a "blunt ended" dsR A is a dsRNA that is double-stranded over its entire length, i.e., no nucleotide overhang at either end of the molecule.
  • antisense strand refers to the strand of a dsRNA which includes a region that Is substantially complementary to a target sequence.
  • region of complementarity refers to the region on the antisense strand, that is substantially complementary to a sequence, for example a target sequence. Where the region of complementarity is not fully complementary to the target sequence, the mismatches are most tolerated outside nucleotides 2-7 of the 5' terminus of the antisense strand
  • sense strand refers to the strand of a dsRNA that includes a region that is substantially complementary to a region of the antisense strand.
  • substantially complementary means preferably at least S5% of the overlapping nucleotides in sense and antisense strand are complementary.
  • Introducing into a cell when referring to a dsRNA, means tacilit ing uptake or absorption into the cell, as is understood by those skilled in the art. Absorption or uptake of dsRNA can occur through unaided diffusive or active cellular processes, or by auxiliary agents or devices, The meaning of this term is not limited to cells in vitro; a dsRNA may also be “introduced into a cell", wherein the ceil is part of a living organism. In such instance, introduction into the cell will include the delivery to the organism.
  • ds&NA can be injected Into a tissue site or administered systemically, it is, for example envisaged that the dsRNA molecules of this invention be administered to a subject in need of medical, intervention.
  • Such an administration may comprise the injection of the dsRNA, the vector or a cell of this invention into a diseased site in said subject, for example into liver tissue/cells or into cancerous tissues cells, like liver cancer tissue.
  • the injection is preferably in close proximity to the diseased tissue envisaged.
  • In vitro introduction into a ceil Includes methods known in the art such as electroporation and lipofeeiion,
  • proliferating and proliferation refer to cells undergoing mitosis.
  • proliferative disorder refers to any disease/disorder marked by unwanted or aberrant proliferation of tissue.
  • proliferative disorder also refers to conditions in which the unregulated and/or abnormal growth of cells can lead to the development of an unwanted condition or disease, which can be cancerous or noncancerous.
  • inflammation refers to the biologic response of body tissue to injury, irritation, or disease which can be caused by harmful stimuli, for example, pathogens, damaged cells, or irritants. Inflammation is typically characterized by pain and swelling. Inflammation is intended to encompass both acute responses, in which inflammatory processes are active (e.g., neutrophils and leukocytes), and chronic responses, which are marked by slow- progress, a shift in the type of cell present at the site of inflammation, and the formation of connective tissue.
  • fibrosis is fibrosis.
  • Cancers to be treated comprise, but are again not limited to leukemia, AML, solid tumors, liver cancer, brain cancer, breast cancer, lung cancer, NSCLC, colorectal cancer, bladder cancer and prostate cancer, whereby said liver cancer may, inter alia, be selected from the group consisting of hepatocellular carcinoma (HCC), hepatoblastoma, a mixed liver cancer, a cancer derived from mesenchymal tissue, a liver sarcoma or a cholangiocarcinoma.
  • HCC hepatocellular carcinoma
  • hepatoblastoma a mixed liver cancer
  • mesenchymal tissue a liver sarcoma
  • a cholangiocarcinoma hepatocellular carcinoma
  • the degree of inhibition is usually expressed in terras of
  • the degree of inhibition may be given in terms of a reduction of a parameter that is functionally linked to the FLT3 gene transcription, e.g. the amount of protein encoded by a FLT3 gene which s secreted by a cell, or the number of cells displaying a certain pherrotype.
  • the inventive dsRNA molecules are capable of inhibiting the expression of a human FLT3 by at least about 60%, preferably by at least 70%, most preferably by at least S0% in vitro assays, Le in vitro.
  • the term "in vitro" as used herein includes but is not limited to cell culture assays.
  • inventive dsRNA molecules are capable of inhibiting the expression of a mouse or rat FLT3 by at least 60 %.preferably by at least 70%, most preferably by at least 80%.
  • the person skilled in the art can readily determine such an inhibition rate and related effects, in particular in light of the assays provided herein.
  • off target refers to all non-target mRKAs of the transcriptorne that are predicted by in silieo methods to hybridize to the described ds ' R As based on sequence complementarity.
  • the dsRNAs of the present invention preferably do specifically inhibit the expression of FLO, i.e. do not Inhibit the expression of any off-target
  • half-Site is a measure of stability of a compound or molecule and can be assessed by methods known to a person skilled in the art, especially in light of the assays provided herein.
  • non-imrnunost!mulatory'' refers to the absence of any induction of a immune response by the invented dsRNA molecules. Methods to determine immune responses are well known to a person skilled in the art for example by assessing the release of cytokines, as described in the examples section,
  • treat means in context of this invention the relief from or aiieviation of a disorder related to FLT3 expression, like inflammation and proliferative disorders, like cancers.
  • a "pharmaceutical composition” comprises a pharmacologically effective amount of a dsRNA and a pharmaceutically acceptable carrier.
  • a “pharmaceutical composition” may also comprise individual strands of such a dsRNA molecule or the herein described veetoris) comprising a regulatory sequence operabiy linked to a nucleotide sequence thai encodes at least one strand of a sense or an antisense strand comprised In the dsRNAs of this invention.
  • cells, tissues or isolated organs that express or comprise the herein defined dsRNAs may be used as "pharmaceutical compositions".
  • “pharmacologically effective amount,” ' ⁇ therapeutically effective amount” or simply “effective amount” refers to thai amount of an RNA effective to produce the intended pharmacological, therapeutic or preventive result.
  • pharmaceutically acceptable carrier refers to a carrier for admin istration of a therapeutic agent
  • Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, etbano!, and combinations thereof.
  • the term specifically excludes cell culture medium.
  • pharmaceutically acceptable carriers include, but are not limited to pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives as known to persons skilled in the art.
  • the pharmaceutically acceptable carrier allows for the systemic administration of the dsRNAs, vectors or cells of this invention.
  • enteric administration is envisaged the parenteral administration and also transdermal or transmueosaS (e.g. insufflation, buccal, vaginal, anal) administration as well as inhalation of the drug are feasible ways of administering to a patient in need of medical intervention the compounds of this invention.
  • parenteral administration can comprise the direct injection of the compounds of this invention into the diseased tissue or at least in close proximity.
  • intravenous, intraarterial subcutaneous, intramuscular, intraperitoneal, intradermal intrathecal and other administrations of the compounds of this invention are within the skill of the artisan, for example the attending physician.
  • compositions of the invention will generally be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pli and Isotonicity.
  • the carrier consists exclusively of an aqueous buffer.
  • exclusively means no auxiliary agents or encapsulating substances are present which might affect or mediate uptake of dsRNA In the cells that express a FLT3 gene.
  • Aqueous suspensions according to the invention may include suspending agents such as cellulose derivatives, sodium alginate, poly vinyl-pyrrol idone and gum tragacanth, and a wetting agent such as lecithin.
  • Suitable preservatives for aqueous suspensions include ethyl and n ⁇ prop i p-hydrox benzoate.
  • the pharmaceutical compositions useful according to the invention also include encapsulated formulations to protect the dsRNA against rapid elimination from the body, such as a controlled release formulation, including Implants and microencapsulated deliver)' systems.
  • a controlled release formulation including Implants and microencapsulated deliver
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and potylactic acid, Methods for preparation of such formulations will be apparent to those skilled in the art.
  • Liposomal suspensions can also be used as pharmaceutically acceptable carriers. These ears be prepared according to methods known to those skilled in the art, for example, as described in PCX publication WO 91/06309 which is incorporated by reference herein.
  • a "transformed cell” is a cell Into which at least one vector has been Introduced from which a dsHNA molecule or at least one strand of such a dsRNA molecule may be expressed.
  • a vector is preferably a vector comprising a regulatory sequence operabiy linked to nucleotide sequence that encodes at least one sense strand or antisense strand of a dsRNA of the present invention.
  • dsRNAs comprising one of the sequences in Table 1 and 4 minus only a few nucleotides on one or both ends may be similarly effective as compared to the dsRNAs described above.
  • inventive dsRNA molecules comprise nucleotides 1-1 of the sequences given in Table 1.
  • the ds MA molecules provided heresn comprise a duplex length (i.e. without “overhangs") of about 16 to about 30 nucleotides, Particular useful dsRNA duplex lengths are about 19 to about 25 nucleotides. Most preferred are duplex structures with a length of 19 nucleotides, in the inventive dsRNA molecules, the antisense strand is at least partially complementary to the sense strand.
  • the dsRNA of the invention can contain one or more mismatches to the target sequence.
  • the dsRNA of the invention contains no more than 13 mismatches, if the antisense strand of the dsRNA contains mismatches to a target sequence, it is preferable that the area of mismatch not be located within nucleotides 2-7 of the 5' terminus of the antisense strand. In another embodiment it is preferable that the area of mismatch not be located within nucleotides 2-9 of the 5' terminus of the antisense strand.
  • ds NAs having at least one nucleotide overhang may have a single-stranded nucleotide overhang of 1 to 5, preferably I or 2 nucleotides
  • ds NAs having at least one nucleotide overhang have unexpectedly superior inhibitory properties than their blunt-ended counterparts.
  • the present inventors have discovered that the presence of only one nucleotide overhang strengthens the interference activity of the dsRNA, without affecting its overall stability. dsRNA having only one overhang has proven particularly stable and effective in vivo, as well as in a variety of cells, cell culture mediums, blood, and serum.
  • the single-stranded overhang is located at the S'-terminal end of the antisense strand or, alternatively, at the S'-term al end of the sense strand.
  • the dsRNA may also have a blunt end, preferably located at the S'-end of the antisense strand.
  • the antisense strand of the dsRNA has a nucleotide overhang at the 3 ? -end, and the 5 '-end is blunt.
  • one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate.
  • the dsRNA of the present invention may also be chemically modified to enhance stability.
  • the nucleic acids of the invention may be synthesized and/or modified by methods well established in the art, such as those described in "Current protocols in nucleic acid chemistry", Beaueage, S.L. et al. (Edrs.), John Wiley & Sons. Inc., New York, NY, USA, which is hereby incorporated herein by reference.
  • Chemical modifications may include, but are not limited to 2' modifications, introduction of non-natural bases, eovalent attachment to a ligand, and replacement of phosphate linkages with thiophosphate linkages, inverted deoxythyniidines, in this embodiment, the integrity of the duplex structure is strengthened by at least one, and preferably two, chemical linkages.
  • Chemical linking may be achieved by any of a variety of well-known techniques, for example by introducing eovalent, ionic or hydrogen bonds; hydrophobic interactions, van der Waais or stacking interactions; by means of metal-ion coordination, or through use of purine analogues
  • the chemical groups that can be used to modify the dsRNA include, without limitation, methylene blue; blfonctional groups, preferably bis-(2-ehloroethyl)annne; N-acetyi-N' ⁇ p-glyoxylbenzoyiJcystamine 4-thiouracii; and psoralen
  • the linker is a hexa-ethylene glycol linker.
  • the dsRNA are produced by solid phase synthesis and the hexa-eihylene glycol linker is incorporated according to standard methods (e.g., Williams, DX, and ,B. Hall, Bioch m. (1996) 35: 14665-14670),
  • the S'-ersd of the antisense strand and the 3'-end of the sense strand are chemically linked via a hexaethylene glycol linker
  • at least one nucleotide of the dsRNA comprises a phosphorothioate or phosphorodlthi shareholders.
  • the chemical bond at the ends of the dsRNA is preferably formed by triple-helk bonds
  • a chemical bond may be formed by means of one or several bonding groups, wherein such bonding groups are preferably poiy-(oxyphosphinicooxy-l ; 3- propandiol)- and/or polyethylene glycol chains.
  • a chemical bond may also be formed by means of purine analogs introduced into the double-stranded structure instead of purines.
  • a chemical bond may be formed by azahen ⁇ ene units introduced into the double-stranded structure.
  • a chemlcai bond may be formed by branched nucleotide analogs instead of nucleotides introduced into the double- stranded structure.
  • a chemical bond may be induced by ultraviolet light.
  • the nucleotides at one or both of the two single strands may be modified to prevent or Inhibit the activation of cellular enzymes, for example certain nucleases.
  • Techniques for inhibiting the activation of cellular enzymes are known in the art including, but not limited to, 2' -amino modifications, 2 ? -amino sugar modifications, 2'-F sugar modifications, 2'-F modifications, 2' ⁇ alkyl sugar modifications, uncharged backbone modifications, morpholino modifications, 2' ⁇ Q ⁇ methy! modifications, and phosphoramidate (see, e.g., Wagner, Nat Med, (1995) 1 : 1 1 16-8).
  • nucleotide group of the nucleotides on a dsRMA is replaced by a chemical group, preferably by a 2 * -amino or a 2'- rnethyl group.
  • at least one nucleotide may be modified to form a locked nucleotide.
  • Such locked nucleotide contains a methylene bridge that connects the 2 * -oxygen of ribose with the 4 s - carbon of ribose.
  • Introduction of a locked nucleotide into an oligonucleotide improves the affinity for complementary sequences and increases the melting temperature by several degrees.
  • Modifications of dsRNA molecules provided herein may positively influence their stability in vivo as well as in vitro and also improve their delivery to the (diseased) target side, Furthermore, such structural and chemical modifications may positively influence physiological reactions towards the dsRNA molecules upon administration, e.g. the cytokine release which is preferably suppressed.
  • Such chemical and structural modifications are known in the ait and are, inter alia, illustrated in Nawrot (2006) Current Topics in Med Chem, 6, 13-925.
  • Conjugating a iigand to a dsRNA can enhance its cellular absorption as well as targeting lo a particular tissue, in certain instances, a hydrophobic ligand is conjugated to the dsRNA to facilitate direct permeation of the cellular membrane.
  • the ligand conjugated to the ds NA is a substrate for receptor-mediated endocytosis.
  • lipophilic compounds that have been conjugated to oligonucleotides include 1-pyrene butyric acid, t3-his-0-(hexadecyi)glyeerol, and menthol.
  • a Hgand for receptor-mediated endocytosis is folic acid. Folic acid enters the cell by toiate-receptor-mediated endocytosis. dsRNA compounds bearing folic acid would be efficiently transported into the cell via the folate-receptor-rnediated endocytosis.
  • oligonucleotide Attachment of folic acid to the 3 '-terminus of an oligonucleotide results in increased cellular uptake of the oligonucleotide (Li, S.; Deshniukh, H. M Press; Huang, L. Ph rm. Res, 1998, 15, 1540).
  • Other ligands that have been conjugated to oligonucleotides include polyethylene glycols, carbohydrate clusters, cross-linking agents, porphyrin conjugates, and delivery peptides. in certain instances, conjugation of a cationic Hgand to oligonucleotides often results in improved resistance to nucleases.
  • cationic ligands are propylammonium and dimethylpropylairimonium.
  • antisense oligonucleotides were reported to retain their high binding affinity to mRNA when the cationic hgand was dispersed throughout the oligonucleotide.
  • the ligand-eonjugated dsRNA of the invention may be synthesized hy the use of a dsR A that bears a pendant reactive functionality, such as that derived from the attachment of a linking molecule onto the dsRMA.
  • This reactive oligonucleotide may be reacted directly with commercially-available ligands, ligands that are synthesized bearing any of a variety of protecting groups, or ligands that have a linking moiety attached thereto.
  • the methods of the invention facilitate the synthesis of ligand-eonjugated dsRNA hy the use of, in some preferred embodiments, nucleoside monomers that have been appropriately conjugated with ligands and that may further be attached to a solid-support material.
  • Such !igand-nucleoside conjugates are prepared according to some preferred embodiments of the methods of the invention via reaction of a selected serum-binding ligand with a linking moiety located on the 5' position of a nucleoside or oligonucleotide, in certain instances, an dsRNA bearing an aralkyl ligand attached to the S'-terminus of the dsRNA is prepared by first covalently attaching a monomer building block to a control ied-pore-glass support via a long-chain arninoaikyi group. Then, nucleotides are bonded via standard solid- phase synthesis techniques to the monomer building-block bound to tJhe solid support.
  • Hie monomer building block may be a nucleoside or other organic compound thai is compatible with solid-phase synthesis.
  • the dsRNA used ire the conjugates of the invention may be conveniently and routinely made through the well-known technique of solid-phase synthesis. It is also known to use similar techniques to prepare other oligonucleotides, such as the phosphorothioates and alkylated derivatives.
  • the oligonucleotides and oligonucieosides may be assembled on a suitable DNA synthesizer utilizing standard nucleotide or nucleoside precursors, or nucleotide or nucleoside conjugate precursors that already bear the linking moiety, ligand-nucleoiide or nucleoside-eonjugate precursors that already hear the ligand molecule, or non-nacleoslde ligaitd- bearing building blocks.
  • nudeotide-eonjugate precursors thai already bear a linking moiety the synthesis of the sequence-specific linked nucleosides is typically completed, and the ligand molecule Is then reacted with the linking moiety to form the ligand-conjugated oligonucleotide.
  • the oligonucleotides or linked nucleosides of the invention are synthesized by an automated synthesizer using phosphoramidites derived from lijgand ⁇ nudeos.de conjugates in addition to commercially available phosphoramidites.
  • functionalized, linked nucleosides of the invention can be augmented to include either or both a phosphorothioate backbone or a 2 -0- methyl, 2'-0-ethyi, 2" ⁇ 0 ⁇ propyl, 2 i -0-aminoaikyi J 2"-0-allyl or 2'-deGxy-2' ⁇ i!uGro group.
  • functional ized nucleoside sequences of the invention possessing an amino group at the S'-terminus are prepared using a DNA synthesizer, and then reacted with an active ester derivative of a selected ligand. Active ester derivatives are well known to those skilled in the art.
  • active esters include N-hydrosuccinimide esters, tetrafluorophenolic esters, pentaflnorophenolic esters and pentachlorophenoiic esters.
  • the reaction of the amino group and the active ester produces an oligonucleotide in which the selected ligand is attached to the S'-position Jurough a linking group.
  • the amino group at the 5' ⁇ terminus can be prepared utilizing a 5'-Am mo-Modifier C6 reagent.
  • ligand molecules may be conjugated to oligonucleotides at the 5 -position by the use of a ligand- nucleoside phosphoramidite wherein the ligand is linked to the 5'-hydroxy group directly or indirectly via a linker.
  • ligand-nucleoside phosphoramidites are typically used at the end of an automated synthesis procedure to provide a Hgand-conjugated oligonucleotide bearing the ligand at the 5' ⁇ terminus.
  • the preparation of ligand conjugated oligonucleotides commences with the selection of appropriate precursor molecules upon which to construct the ligand molecule.
  • die precursor is an appropriately- protected derivative of the eommonSy-used nucleosides.
  • the synthetic precursors for the synthesis of the ligand-conjugated oligonucleotides of the invention include, but are not limited to, 2'-ammoaikoxy ⁇ 5 t -ODMT-n «cSeosides s , 2 , -6-aniinoaikyla ino-5'-GDMT-nueleosides, S'-S-aminoaikoxy- ⁇ -deoxy-nueleosides, 5'-6 ⁇ aminoaIkoxy-2-proteeted-niscieosides, 3 -6- amsnoalkoxy-5'-ODMT-nucleosides, and S'-aminoalkyiamiriO-S'-ODMT-rsucIeosidos that may be protected in the nucleohase portion of the molecule.
  • Methods for the synthesis of such amino- linked protected nucleoside precursors are know to those of ordinary skill in the art.
  • protecting groups are used during the preparation of the compounds of the invention.
  • the term "protected” means that the indicated moiety has a protecting group appended thereon.
  • compounds contain one or more protecting groups.
  • protecting groups can be employed in the methods of the invention, in general, protecting groups render chemicai functionalities inert to specific reaction conditions, and can be appended to and removed from such functionalities in a molecule without substantially damaging the remainder of the molecule.
  • Amino-protecting groups stable to acid treatment are selectively removed with base treatment and are used to make reactive amino groups selectively available for substitution.
  • Examples of such groups are the Fmoc (E. Atherton and R. C. Sheppard in The Peptides, S. Udenfriersd, J. Meienhofer, Eds. s Academic Press, Orlando, 1987, volume 9, p,l) and various substituted sulfonyiethy! carbamates exemplified by the Nsc group (Samukov et al. Tetrahedron LetL, 1994, 35:7821.
  • Additional amino-protecting groups include, but are not limited to, carbamate protecting groups, such as 2 rhnethylsilylethoxycarbon I (Teoc), 1 -methyl- 1 - ⁇ 4- biphenylyi)ethoxycarbonyS (Bpoc), t-butoxycarbonyi (BOG), allyloxycarbonyS (Alloc), 9 ⁇ fluorenylmethyloxyearbonyl (Fmoc), and benzy!oxycarbonyl (Cbz); amide protecting groups, such as formyl, acetyl trihaloaeetyl, benzoyl, and mtrophenylaeetyS; sulfonamide protecting groups, such as 2-nitrobenzenesu!fonyl; and imine and cyclic imide protecting groups, such as phthalimido and diihiasuecmoyt Equivalents of these amino-protecting groups are also encompassed by the compounds and methods of the invention, Many solid supports
  • a universal support allows for the preparation of oligonucleotides having unusual or modified nucleotides located at the 3 5 erminus of the oligonucleotide.
  • For further details about universal supports see Scott et ah, JnttovaHom and Perspectives in solid-phase Synthesis, 3rd International Symposium, 1.994, Ed. Roger Epton, Mayflower Worldwide, 115-124].
  • the nucleosides are linked by phosphorus-containing or non-phosphorus-eontaining covaleut intemueleoside linkages,
  • conjugated nucleosides can be characterized as ligand-bearing nucleosides or ligand-nucieoside conjugates.
  • the linked nucleosides having an aralkyl ligand conjugated to a nucleoside within their sequence will demonstrate enhanced dsRNA activity when compared to like ds NA compounds that are not conjugated.
  • the araikyS-iigand-eonjugated oligonucleotides of the invention also include conjugates of oligonucleotides and linked nucleosides wherein the ligand is attached directly to the nucleoside or nucleotide without the intermediacy of a linker group.
  • the ligand may preferably he attached, via linking groups., at a carboxyl, amino or oxo group of the ligand.
  • Typical linking groups may be ester, amide or carbamate groups.
  • modified oligonucleotides envisioned for use in the ligand-conjugated oligonucleotides of the invention include oligonucleotides containing modified backbones or non-natural internucieoside linkages.
  • oligonucleotides having modified backbones or intemueleoside linkages include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone.
  • modified oligonucleotides that do not have a phosphorus atom in their intersugar backbone can also be considered to be oligonucieosides
  • Specific oligonucleotide chemical modifications are described below. It is not necessary for all positions in a given compound to be uniformly modified. Conversely, more than one modifications may be incorporated in a single dsRNA compound or even in a single nucleotide thereof.
  • Preferred modified intemucleoside linkages or backbones include, for example, phosphorothioates, ehiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalk iphosphotriesters, methyl and other alky! phosphonates including 3'-a!k lene phospSionates and chiral phosphonates, phosphinates, phosphoramidates including 3' ⁇ ammo phosphoraniidate and ammoalkylphosphoramidates, thionophosphoramidates, thionoaikyiphosphonaies, thionoalkylphosphotriesters, and boranophosp ates having normal 3 !
  • Representative United States Patents relating to the preparation of the above phosphorus- atom-containing linkages include, but are not limited to, L S. Pat. Nos. 4,469,863; 5,023,243; 5,264,423; 5,321 , 131 ; 5,399,676; 5,405,939; 5,453,496; 5,455,233 and 5,466,677, each of which is herein incorporated hy reference.
  • Preferred modified intemucleoside linkages or backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycioalkyl intersugar linkages, mixed heteroatom and aikyl or cycioalkyl iniersugar linkages, or one or more short chain heteroatomk or heterocyclic intersugar linkages. These include those having morphoHrro linkages ⁇ formed in part from the sugar portion of a nucleoside); slloxane backbones; sulfide, sulfoxide and sulfone backbones; formaeety!
  • PNA peptide nucleic acid
  • Some preferred embodiments of the invention employ oligonucleotides with phosphorothioate linkages and oligonucleotides with heteroatorn backbones, and in particular— CH2--NH--O-CH2 --, ⁇ CH2TMN(CIl3)TM0-C!I 2 - [known as a methylene (methyli ino) or MM! backbone], -CH 2 -0-N(CH 3 ) ⁇ CH 2 ⁇ CH2-N(CH3)- (CH 3 ) ⁇ CH2-, and -0TMNiC3 ⁇ 4)TMCH 2 --C3 ⁇ 4— [wherein the native phosphodiester backbone is represented as ⁇ — p— 0— CF3 ⁇ 4 ⁇ ] of the above referenced U.S.
  • oligonucleotides employed in the ligand-conjugated oligonucleotides of the invention may additionally or alternatively comprise nucleobase (often referred to in the art simply as “base”) modifications or substitutions.
  • nucleobase often referred to in the art simply as “base”
  • “unmodified” or “natural” nucieobases include the purine bases adenine (A) and guanine (G), and the pyrlmidine bases thymine (T), cytosine (C), and uracil (U).
  • Modified nucieobases include other synthetic and natural nucieobases, such as 5 ⁇ methy!eyiosine (5 ⁇ nie ⁇ C) s 5-hydroxymeihyi cytosine, xanthine, hypoxanthine, 2 ⁇ aminoadenine, 6 ⁇ methyl and other aikyl derivatives of adenine and guanine, 2- propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2- thiocytossne, 5-halouracil and cytosine, 5-propynyI uracil arid cytosine, 6-azo uracil, cytosine and thymine, 5-uracsI (pseudouracii), 4-thiouracil, 8-haio, 8-aniino s 8-thiol, 8-ihioaikyl, 8- hydroxyl and other 8 ⁇ substituted
  • nucleobases include those disclosed in U.S. Pat, No. 3,687,808, those disclosed in the Concise Encyclopedia Of Poiymer Science And Engineering, pages ⁇ 58-859, Kroschwite, J. L, ed, John Wiley & Sons, 1990, those disclosed by Engiisch et al. s Ange a ite Chemie. Internationa! Edition, 1991 , 30, 613, and tbose disclosed by Sanghvi, Y, S neutral, Chapter 15, A isense Research an Applications, pages 289-302, Crooke, S. T, and Lebleu, B.noc ed., CRC Press, 1993.
  • nucleobases are particularly useful for increasing the binding affinity of the oligonucleotides of the invention.
  • These include 5-sufastituted pyrimidines, 6- azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-antinopropyladenine, 5- propynyluracil and S-propynyicytosine.
  • S-Methyleytosme substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2 °C. (Id., pages 276-278) and are presently preferred base substitutions, even more particularly when combined with 2 -methoxyethyt sugar modifications.
  • oligonucleotides employed in the Hgand-eonjugated oligonucleotides of the invention may additionally or alternatively comprise one or more substituted sugar moieties.
  • Preferred oligonucleotides comprise one of the following at the 2" position: OH: F; 0 ⁇ , S-, or N-alkyl, G-, S ⁇ , or N-alkenyl, or O, S ⁇ or M ⁇ aikynyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted Cj to Cio alky! or C to Cjo alkeny! and alkynyl.
  • n and ni are from 1 to about 10.
  • oligonucleotides comprise one of the following at the 2 * position: Q to Cjo lower alkyl, substituted lower alkyl, alkaryi, aralkyt 0 ⁇ alkaryi or O-araiky!, SH, SCH 3 , OCN, CI, Br, C , CF 3> OCF 3) SOCH 3 , S0 2 CH 3 , ON0 2 , NO 3 ⁇ 4 N 3> NH 2 ⁇ heterocydoalkyl heterocycloalkary!, am oalkylamino, pofyaikylamino, substituted silyi, an NA cleaving group, a reporter group, an Inierealator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving die pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties.
  • a preferred modification includes 2 -methoxyethoxy also known as 2'-G-(2 ⁇ methoxyethyi) or 2'- MOE], i.e., an alkoxyalkoxy group.
  • a further preferred modification includes 2'- dimethylaminooxyethoxy, i.e., a 0 ⁇ €3 ⁇ 4) 2 ⁇ 3 ⁇ 4) 2 group, also known as 2'-DMAOE, as described in U.S. Pat. No. 6, 127,533, filed on Jan. 30, 1998, the contents of which are incorporated by reference.
  • sugar substituent group or "2'-subsi!tueni group” includes groups attached to the mposition of the nbofiiranosyl moiety with or without an oxygen atom
  • Sugar substituent groups include, but are not limited to, fluoro, Q-alky!, O-alkylamino, O- alkylaikoxy, protected O-alkylamino, O-afkylar noalkyi, Oaiky! imidazole and polyethers of the formula (0-alkyl) ro , wherein rn is 1 to about 10.
  • polyethers are iinear and cyclic polyethylene glycols (PEGs), and (PEG)-containing groups, such as crown ethers and, inter alia, those which are disclosed by Delgardo et. ai. (Critical Reviews in Therapeutic Drug Carrier Systems S 992, 9:249), which is hereby incorporated by reference in its entirety. Further sugar modifications are disclosed by Cook ⁇ A i-fibrosts Drug Design, 1991 , 6:585-607), Fluoro, Q-alkyl, Q ⁇ atkylamino, O-alkyl imidazole, G-aikylarninoalkyl, and aSkyl amino substitution is described in U.S.
  • Patent 6, 166, 397 entitled "Oligomeric Compounds having Pyrimidine NucSeotide(s) with 2' and 5' Substitutions," hereby incorporated by reference in its entirety.
  • Additional sugar substituent groups amenable to the invention include 2 -S and 2 - R2 groups, wherein each R is, independently, hydrogen, a protecting group or substituted or unsubststuted alkyl, alkenyk or alkynyh 2 -S Nucleosides are disclosed in U.S. Pat. No, 5,670,633, hereby incorporated by reference in its entirety.
  • the incorporation of 2 -SR monomer synthons is disclosed by Hamm et al. (J.
  • 2 -NR nucleosides are disclosed by Goett gers, M, J, Org. Chenh, 1996, 61, 6273-6281 ; and Polushin et al. Tetrahedron Lett., 1 96, 37, 3227-3230, Further representative 2 -substituent groups amenable to the invention include those having one of formula I or II:
  • E is ⁇ 3 ⁇ 4)( 3 ⁇ 4); each % and Q4 is.
  • q 3 is an integer from i to 1 ;
  • 3 ⁇ 4 Is an integer from 1 to 10;
  • each Z Z2 arid Z3 is, independently, C4-C7 eycloaikyl, aryi or C Qs eterocyclyi, wherein the heteroatom in said heterocyclyl group is selected from oxygen, nitrogen and sulfur;
  • Z 4 is OMj, S 3> or N(Mj) 2 5 each M ⁇ is, independently. H, C ⁇ -(1 ⁇ 4 alkyi, C 3 -Cs haloalkyl, C( » NH)N(H)M 2 , C(0)N(H)M 2 or OC( ))NiH)M 2 ; M 2 is H or C Cg alkyl; and
  • Zj is Ct-C- alkyl Cj -CJO haioalkyi, C2-C10 alkenyl, C2-CK1 alkynyl, €3 ⁇ 4-Cs aryi, ( ⁇ 1 ⁇ 4)( ⁇ & . ), OQ 3 , halo, SQ 3 or CN.
  • Representative 2'-0-sugsr substituent groups of formula I are disclosed in U.S. Pat No, 6,172,209, eruilled "Capped 2 -Oxyethoxy Oligonucleotides,” hereby incorporated by reference in its entirety.
  • Representative cyclic 2 '-O-sugar substituent groups of formula II are disclosed in U.S. Patent 6,271,358, entitled “UNA Targeted 2'-Madified Oligonucleotides that are Conforrnationaliy Preorganized,” hereby incorporated by reference in its entirety.
  • Sugars having O-suhstitutions on the ribosyl ring are also amenable to the invention.
  • Representative substitutions for ring O include, but are not limited to, S, C3 ⁇ 4, CHF, and CF 2 ,
  • Oligonucleotides may also have sugar ndrnetics, such as cydohutyi moieties, In place of the pentofuraraosyl sugar.
  • sugar ndrnetics such as cydohutyi moieties
  • Representative United States patents relating to the preparation of such modified sugars include, but are not limited to, U.S. Pat, Nos, 5,359,044; 5,466,? ⁇ 6; 5,51 , 134; 5,591,722; 5,597,909; 5,646,265 and 5,700,920, ail of which are hereby incorporated by reference.
  • iigand-conjugated oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more additional non ⁇ iigand moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide.
  • moieties include hut are not limited to lipid moieties, such as a cholesterol moiety (Letsinger et aL, Froc. Natl. Acad. Sci.
  • compositions employing oligonucleotides that are substantially chiraiiy pure with regard to particular positions within the oligonucleotides include, but are not limited to, those having phosphorothioate linkages that are at least 75% Sp or Rp (Cook et a!., U.S. Pat. No. 5,587,361) and those having substantially chiraiiy pure (Sp or Rp) alkyiphosphonate, phosphoramldate or phosphoiriester linkages (Cook, U.S. Pat Nos. 5.212,295 and 5,521,302).
  • the oligonucleotide may be modified by a non-ligand group.
  • a number of non-Hgand molecules have been conjugated to oligonucleotides in order to enhance the activity, cellular distribution or cellular uptake of the oligonucleotide, and procedures for performing such conjugations are available in the scientific literature.
  • Such non-Hgand moieties have included lipid moieties, such as cholesterol (Letsinger et at., Proc. Nad. Acad. Sci USA, 1989, 86:6553), choiie acid (Manoharan et a!,, Bioorg, Med. Chem.
  • a thioether e.g., hexyl-S-trityithioS (Manoharan et al, Aim. NY. Acad Sci., 1992, 660:306; Manoharan et at., Bioorg. Med Chem. Iei.
  • a phospholipid e.g., di-hexadecyl-rac-glyeerol or triethylammonium l,2-di-0-hexadeeyl-rac-g!yeero-3-H-phosphonate (Manoharan et al, Tetrahedron Lett, 1995, 36:3651 ; Shea et al., Nnd.
  • Typical conjugation protocols involve the synthesis of oligonucleotides bearing an aminolinker at one or more positions of the sequence. The amino group is then reacted with the molecule being conjugated using appropriate coupling or activating reagents. The conjugation reaction may be performed either with the oligonucleotide still hound to the solid support or foliowing cleavage of the oligonucleotide in solution phase. Purification of the oligonucleotide conjugate by BPLC typically affords the pure conjugate.
  • the molecule being conjugated may be converted into a building block, such as a phosphora idite, via an alcohol group present in the molecule or by attachment of a linker bearing an alcohol group that tnay be phosphory!ated.
  • a building block such as a phosphora idite
  • each of these approaches may be used for the synthesis of ligand conjugated oligonucleotides.
  • Amino linked oligonucleotides may be coupled directly with ligand via the use of coupling reagents or foliowing activation of the ligand as an NHS or pentfluorophenolate ester.
  • Ligand phosphorarnidites may be synthesized via tbe attachment of an aminohexanol linker to one of the carboxyl groups followed by phosphitylation of the terminal alcohol functionality.
  • Other linkers, such as cysteamine may also be utilized for conjugation to a chioroacetyl linker present on a synthesized oligonucleotide.
  • nucleic acid molecules or the vectors of this invention, encoding for at least one strand of the inventive dsKNAs may be introduced into cells or tissues by methods known in the art, like transfeetions etc.
  • dsRNA molecules for the introduction of dsRNA molecules, means and methods have been provided.
  • targeted delivery by glycosylated and folate-modifsed molecules including the use of polymeric carriers with ligands, such as galactose and lactose or the attachment of folic acid to various maeromoieeules allows the binding of molecules to be delivered to folate receptors.
  • Targeted delivery by peptides and proteins other than antibodies for example, including GD- modified nanoparttcies to deliver siRNA in vivo or multieonsponent (nonvlral) delivery systems including short eyelodextrins, adamaniine-PEG are known, Yet, also the targeted delivery using antibodies or antibody fragments, incliiding (monovalent) Fab-fragments of an antibody (or other fragments of such an antibody) or single-chain antibodies are envisaged, injection approaches for target directed deliver)' comprise, inter alia, hydrodynamlc i,v. injection.
  • cholesterol conjugates of dsRNA may be used for targeted delivery, whereby the conjugation to lipohilte groups enhances ceil uptake and improve pharmacokinetics and tissue biodlstribution of oligonucleotides.
  • cationic delivery systems are known, whereby synthetic vectors with net positive (cationic) charge to facilitate the complex formation with the polyanionic nucleic acid and interaction with the negatively charged cell membrane.
  • Such cationic deliver)' systems comprise also cationic liposomal delivery systems, cationic polymer and peptide delivery systems.
  • Other delivery systems for the cellular uptake of dsRNA/siR A are aptamer-ds siRNA.
  • gene therapy approaches can he used to deliver the inventive dsRNA molecules or nucleic acid molecules encoding the same.
  • Such systems comprise the use of non-pathogenic vires, modified viral vectors, as well as deliveries with nanoparticles or liposomes.
  • Other delivery methods for the cellular uptake of dsRNA are extracorporeal, for example ex vivo treatments of cells, organs or tissues, Certain of these technologies are described and summarized in publications, like Akhtat (2007), Journal of Clinical investigation 1 17, 3623*3632, Nguyen ei ⁇ (2008), Current Opinion in Moleculare Therapeutics 10, 138-167. Zamhoni (2005), Clin Cancer Res 1 1, 8230-8234 or Ikeda ei ⁇ .
  • Table 1 Core sequences of dsRNAs targeting human FLT3 gene Letters in capitals represent RNA nucleotides.
  • Table 3 -Characterization of dsRNAs targeting human FLT3: Activity testing far dose response. IC 50; 50 % inhibitory concentration, 1C SO: SO % inhibitory concentration, 1C 20: 20 % inhibitor)' concentration.
  • Table 4 Characterization of dsRNAs targeting human FLT3: Stability and Cytokine Induction, t 1 ⁇ 2 : half-life of a strand as defined in examples, PBMC; Human peripheral blood mononuclear cells. able 5 - Core sequences of dsRNAs targeting human FLT3 gene and their modified counterparts. Letters in capitals represent RNA nucleotides, lower ease letters “c 55 , "g", "a” and “u” represent 2' O-methyl-niodif ed nucleotides, "s” represents phosphorothioate and "dT” deoxythyrnldine.
  • Table 10 Selected off-targets of dsRNA targeting FLT3 ? SEQ ID NO pair 293/294.
  • Table 11 Sequences of bDNA probes for determination of reduction in off-target expression by dsRNA SEQ ID NO pair 295/296.
  • d$RN A design was carried out to identify dsRNAs specifically targeting human FLT3 for therapeutic use.
  • the known mRNA sequence of human (Homo sapiens) FLT3 (NM_O041 19,2 listed as SEQ ID NO. 1 50) was downloaded from NCBI Genbank.
  • the eynomolgous monkey (Macaea fascicularis) FLT3 gene was sequenced (see SEQ ID NO. 1622)
  • the cyrsomolgus monkey sequence (SEQ ID NO. 1651) was examined together with the human FLT3 mRNA sequence (SEQ ID NO, 1650) by computer analysis to identify homologous sequences of 19 nucleotides that yield RNA interference (RNAi) agents eross-reaetive to both sequences.
  • RNAi RNA interference
  • RNAi agents In identifying RNAi agents, the selection was limited to 19mer sequences having at least 2 mismatches lo any other sequence in the human RefSeq database (release 3S ⁇ ⁇ which we assumed to represent the comprehensive human iranseriptome, by using a proprietary algorithm. All sequences containing 4 or more consecutive G ' s (poly-G sequences) were excluded from the synthesis,
  • RNAi agents formed the basis for the synthesis of the RNAi agents in appended Tables 1, 2 and 5.
  • dsRNAs cross-reactive to human as well as cynomoigoiis monkey were defined as most preferable for therapeutic use.
  • such reagent may be obtained from any supplier of reagents for molecular biology at a quality/purity standard for application Irs molecular biology.
  • Oligoribonucleotides were assembled on an AB1 3900 synthesizer (Applied Biosystems) according to the phosphoramsdite oiigornerlzation chemistry.
  • the solid support was polystyrene loaded with 2' ⁇ deoxy ⁇ thyrnldine (purchased from Glen Research, Sterling s Virginia, USA) to give a synthesis scale of 0.2 ⁇ .
  • Ancillary synthesis reagents, RNA and 2'- -Methy! NA phosphoramidiies were obtained from SA C Proilgo (Hamburg, Germany).
  • Coupling time for ail phosphoramidites was 3 min employing 5-EthylthIo-I H- tetrazole (ETT) as activator (0.5 M In Acetonitriie), Phosphorothloate linkages were Introduced using 50 mM 3 (DImethySaniIno ⁇ methylideiie)amIno)-3H-l s 2 f 4-dithIazoie ⁇ 3-thione (DDTT, AM Chemicals, Oceanside, California, USA) in a 1 ; 1 (v/v) mixture of pyridine and Acetonitriie, Upon completion of the solid phase synthesis oHgorlbonucleotides were cleaved from the solid support and deprotected using slight modification of published methods (WincottF.
  • ETT 5-EthylthIo-I H- tetrazole
  • oligoribonucieotldes were purified by anion- exchange high-performance liquid chromatography (HPLC) on a AKTA Explorer System (GF. Healthcare, Freiburg, Germany), Purified oiigorihonucleotldes were desalted by size exclusion chromatography employing a HiTrap 5 niL column (GE Healthcare).
  • oligoribomseleotides were confirmed by MALDi mass spectrometry and purity was assessed by 5 analytical anion-exchange HPLC, To generate siRNAs from RNA single strands, equimolar amounts of complementary sense and antisense strands were mixed and annealed in a 20 mM NaCi, 4 mM sodium phosphate pH 6,8 btrf er, siRNAs were further characterized by capillary gel electrophoresis and were stored f oz until use.
  • MOLM13 ceils were obtained from DSAfZ Deutsche Sammlung von Mikroorganlsmen und Zelikulturen GmbH (Braunschweig, Germany, cat No, ACC-554) and 0 cultured in suspension in RP I 1640 (Bioehrorn AG, Berlin, Germany, cat. No FG 1215)
  • FCS fetal calf serum
  • Penicillin / Streptomycin Gibco Invitrogen, cat. No.15140-122
  • COS7 cells were obtained from DSMZ Deutsche Sammlung 5 von Mikroorganismen und Zelikulturen GmbH (Braunschweig, Germany, cat No. ACC-60) and cultured m DMEM (Biochrom AG, Berlin, Germany, cat. No.
  • F0435 supplemented to contain 10% fetal calf serum (FCS) (Gibco Inviirogen catNo.16250-078), 1% Penicillin / Streptomycin (Gibco Invitrogen, cat No.15140- 122), 5 ml L-Glutamin (Biochrom AG, Berlin, Germany, cat No 0283) and 5 mi 1 ,2 mg mi Natriumbicarbonat (Biochrom AG, Berlin, Germany, cat No L0 1703) at 3 C in an atmosphere with 5% C02 in a humidified incubator (Heraeus HERAceil, endro Laboratory Products, Langenselbold, Germany), MCF7 cells were obtained from DSMZ Deutsche Sammlung von Mikroorganismen und Zelikulturen GmbH (Braunschweig, Germany, cat No.
  • FCS fetal calf serum
  • Penicillin / Streptomycin Gibco Invitrogen, cat No.15140- 122
  • 5 ml L-Glutamin Biochrom AG, Berlin
  • ACC 1 15) and cultured Irs RPMI 1640 (Biochrom AG, Berlin, Germany, cat. No. FG 1215) su lemented to contain 10% fetal calf serum (FCS) (Gibco Invitrogen catNo.16250-078), 1% Penicillin / Streptomycin (Gibco nvitrogen, cat No.15140-122), 5 ni3/50GmI EAA (nonessential aminoacids) (Biochrom AG, Berlin, Germany, cat No 0293), 5 ml/SGOml
  • FCS fetal calf serum
  • Penicillin / Streptomycin Gibco nvitrogen, cat No.15140-122
  • 5 ni3/50GmI EAA nonessential aminoacids
  • TC71 cells were obtained from DSMZ Deutsche Sammlimg von Mikroorganismen und Zellkiiituren GmbH (Braunschweig, Germany, cat. No, ACC516) and cultured in I DM (Gibco Invitrogen, Germany, cat. No. 21980-032) supplemented to contain 10% fetal calf serum (FCS) (Gibco inviirogen cat.No.16250-078), and 5ml/500mS Penicillin / Streptomycin (Gibco Inviirogen, cat. No.15140-122), at 37 in an atmosphere with 5% CO! in a humidified incubator (Heraeus HERAcell, Kendro Laboratory Products, Langenselboid, Germany).
  • FCS fetal calf serum
  • Penicillin / Streptomycin Gibco Inviirogen, cat. No.15140-122
  • MV4-1 1 ceils were obtained from DSMZ Deutsche Sammlung von Mikroorganismen und ZeHkulturen Grnbl l (Braunschweig, Germany, cat No. ACC 102) and cultured sn RPMI 1640 (Biochrom AG ? Berlin, Germany, cat, No. FG 1215) supplemented to contain 10% fetal calf serum (FCS) (Gibco Invitrogen catNo.16250-078), and 5ml/50Qml Penicillin / Streptomycin (Gibco Invitrogen, cat. No.15140-122), at 37 a C in an atmosphere with 5% C02 in a humidified incubator (Heraeus HERAcell Kendro Laboratory Products, Langenselboid, Germany).
  • FCS fetal calf serum
  • Penicillin / Streptomycin Gibco Invitrogen, cat. No.15140-122
  • ChemQiumineseence was measured in a YIctor2-Light (Perkin Elmer, Wiesbaden, Germany) as RLUs (relative light units) and values obtained with the human KIF10 probeset were normalized to the respective human GAPDH values for each well Unrelated control siRNAs were used as negative control,
  • DualG!o-Reporter assay This assay was performed in a 96-weil plate formal.
  • Lipofectamine 2000 (Invitrogen GmbH, Düsseldorf, Germany, cat.No. 11668-01 was used as transfection reagent following the protocol of the manufacturer. Ceil seeding and iransfection of plasmid-DNA (SOng well) were performed at the same time. Transfection of siRNA was carried out 4 h later. For transfection COS7 cells were seeded at a density of 2.5 times 10 reliesup,4 cells/well. In a first single dose experiment siRNAs were trausfeeted at a concentration of 50 nM. Most effective siRNAs against FLT3 from the single dose screens at 50n were further characterized by dose response curves.
  • Luminescence was measured in a Vietor2 ⁇ Light (Perkin Elmer, Wiesbaden, Germany) as RLUs (relative light units) values obtained for RenIHa Luciferase were normalised to the Firefly Luciferase values for each well Unrelated control siRNAs were used as a negative control.
  • MCF7 cells Cell seeding and transfection of siRNA were performed at the same time. For transfection with siRNA, cells were seeded at a density of 2.5 times 10 sup.4 cells/well in 96-well plates. Transfection of siRNA was carried out with lipofectamine 2000 (.Invitrogen GmbH, Düsseldorf, Germany, ca .No. 1 1668-019) as described by the manufacturer. In a first single dose experiment siRNAs were transacted at a
  • siRNA (nM) 24, 6, 1.5, 0.375, 0.0938, 0.0234, 0.0059, 0.0015, 0.0004 and 0.0001 nM . After transfeetion ceils were incubated at 37*C and 5 %
  • RNA quantification using RT-PCR Total RNA was purified using Qiagen RNeas Kit (Qiagen, Hilden, Germany, cat, No 7 106) following the manufacturer ' s protocol Relative quantification of FLT3 and 18S ribosomal mRNA was carried out with High Capacity cDNA Reverse Transcription Kit ⁇ Applied Biosystems) followed by Taqrnan Gene Expression Assays using the manufacturer's protocol, (Applied Biosystems).
  • Stability of dsRNAs targeting human FLT3 was determined in in vitro assays with either human or mouse serum by measuring the half-life of each single strand. Measurements were carried out in triplicates for each time point, using 3 ⁇ 50 ⁇ dsR A sample mixed with 30 ⁇ 1 human serum (Sigma) or mouse serum (Sigma). Mixtures were incubated for either Gmin, 3 Grain, Ih, 3h, 6h, 24hhong or 4Sh at 37*C. As control for unspecific degradation dsR A was incubated with 30 ⁇ 1 Ix PBS pH 6.8 for 48h. Reactions were stopped by the addition of 4 ⁇ 1 proteinase K (20rng/ml), 25 ⁇ !
  • cytokine induction of dsRNAs was determined by measuring the release of !NF-a and TNF-a in an in vitro PBMC assay.
  • Human peripheral blood mononuclear ceils (PBMC) were isolated from buffy coat blood of two donors by Ficoil eentrifugation at the day of transfection. Cells were transfected in quadruplicates with dsRJNA and cultured for 24h at 37 C C at a final concentration of DQrsM in Opts-MEM, using either Gene Porter 2 (GP2) or DDTAP.
  • the human genome was searched by computer analysis for sequences homologous to the inventive dsRNAs. Homologous sequences that displayed less than 6 mismatches with the inventive dsRNAs were defined as a possible off-targets. Off-targets selected for in vitro off target analysis are given in appended tables g-iO.
  • the human cancer cell line MOLM 13 (DSMZ, Braunschweig, Germany) was maintained in media supplemented with 10% heat-inactivated Fetal Bovine Serum (HI-FBS; GlBCO BRlL Gaithersburg, MD) and 2 mM L-glutamine (GIBCO/BRL),
  • RNA from all collected samples was purified using Qiagen RNeasy Kit following the manufacturer s protocol
  • Relative quantification of FLT3 mRNA and 18S ribosoraal RNA gene expression was carried out with cDNA Reverse Transcription Reagents from Applied Biosystems followed by Taqman Gene Expression Assays (Applied Biosystems) using the manufacturer's protocol.
  • the catalog numbers for each probe set were: human FLT3 (Hs00975659_ml) and 18S (431941 E).
  • Protein concentration were measured by BCA protein assay kit (Therrno Fisher), Equal amount of protein samples was resolved by SDS-polyacr lamide gel electrophoresis using a 4-20% Tris-giyeine gel (Invitrogen) and electroblotted onto a 0.45 ⁇ 3 ⁇ 4 nitrocellulose membrane (invitrogen). Membranes were blocked S hr at room temperature in blocking buffer (5% milk in PBS/0.1% Tween 20) followed by incubation with the primary antibody at 4°C overnight.
  • Results are shown in Figures 6-9.
  • Transfeetion of the Mofm-I 3 AML tumor derived ceil line with siRNAs directed towards FLT3 mRNA produced potent mRNA knockdown which correlated with protein knockdown.
  • the phenofype associated with the loss of FLT3 expression is characterized by the loss of STAT5-F, a Gi arrest producing a swb-Gl population of cells and loss of cell viability.
  • FLT36 CE cagctttgcacctiatccataagTTTTTctcttggaaagaaagt 325
  • FLT326 CE cagcgtgaacatmggtaaattTTTn ctcttggaaagaaagi 345
  • hsGAP002 CE ggagggatetegctcctggaTTTTTetcttggaaagaaagi 310 hsGAP003 CE ccccagcc tctccatggiTTTTTctcttggaaagaaagi 311 hsGAP004 CE gctcccctgcaaigagTTTTTctctiggaaagaaagt 312 hsGAP005 LE agcettgaeggtgccaigTTTTTaggcataggaceegtgtet 313 hsGAP006 LE gatgacaagcttcccgttctcTTTTTaggcaiaggaccegigtci 314 hsGAP0O7 LE agatggigatgggatttccattTTTTTaggcataggacccgtgtcl 315 hsGAPOOS LE gcaicgccccactttt
  • OFP-6 MM 007027,3 TOPBP1 $ AAUUUAAAGAUGAGUACCU 13 14 17 19 nrm.
  • RNFI49 (RNF149), rnRJNA U A ACC AGO U UUCUUGAUUA 13 18 19
  • DRI cofactor2
  • EPB4IL4A20 CE ciccattttccatgtitgcagtTTTTTctctiggaaagaaagt 76S
  • EPB41 L4A23 CE ggciat Ltteattet ilgcitlc fXTTTTe lcttggaaagaaagt 769
  • EPB41L4A27 CE tg gatcacigggagaattgt ' i rrri ' ctcttggaaagaaagt 770
  • EPB41L4A1 LE caa aacaccaaccggag aaiTTTTTgaagtiaccgtitt 771
  • EPB41 L4A12 E tgcltataacgi.aiggatccaaaeTTTTTgaagitaccgtttt 775
  • EPB41L4A16 I LE itcitgtcacattctgaicaggcTTTTTgaagttaccgtt 777
  • EPB4IL4A18 LE gctggctgtgtitgigctattc ' ri ' i ' ri gaagt:.accgtttt 779
  • EPB41L4A19 LE iaicctactgaigctgtitgaltcaTTTTTctgagtcaaagcai 780
  • EPB41L4A21 LE caataattttaattgttccticatral IITgaagttaccgtm 781
  • EPB41L4A22 EE iaaagcittttaciggtgaaggtgTTTTTcigagtcaaagcat 782
  • EPB4IL4A25 LE atittcttcccacggigcafiTi ' TTcigag caaagcat 784
  • EPB41L4A29 LE g ti c ggc gacgcgtgTTTTTetgaglcaaageal 786

Abstract

A pharmaceutical composition comprising a double-stranded ribonucleic acid (dsRNA) for inhibiting the expression of a FLT3 gene, further comprising a pharmaceutically acceptable carrier is disclosed. Also provided are methods for treating diseases caused by the expression of a FLT3 gene using said pharmaceutical composition; and methods for inhibiting the expression of FLT3 in a cell.

Description

Co&s positions asd snefhods for isMMt!ng expression of FLT3 gei¾e§
This invention relates to double-stranded ribonucleic acids (dsRNAs), and their use in mediating RKA Interference to Inhibit the expression of the FLT3 gene. Furthermore, the use of said dsRNA to treat inflammation and proliferative disorders, like cancers, is part of this invention. Cancer remains an important area of high unmet medical need. The majority of current treatments provide small gains in overall survival requiring a delicate balance between efficacy and toxicity. Cancer is characterized by uncontrolled growth and survival driven by the improper regulation of proliferation. The frns~Hke receptor tyrosine kinase-3 (FLT3) plays a key role in the differentiation and growth of immature hematopoietic stem cells and immune cells. Activating mutations in the FLT3 gene create a protein that is constitulively active relaying a constant pro-proSiferative signal in mutant leukemia cells. The presence of such activating mutations is associated with a poor clinical prognosis as well.
Despite significant advances in the field of RNAi and advances in the treatment of fibrosis and proliferative disorders, like cancers, there remains a need tor an agent thai can selectively and efficiently silence the FLT3 gene. A specific FLT3 siRNA is expected to reduce the level of FLT3 mRNA leading to a loss in protein and elimination of STATS phosphorylation. This will produced a loss of growth and an increase in cell death in FLT3 mutant expressing leukemia cells and a reversible growth arrest in wild-type expressing ceils,
The invention provides double-stranded ribonucleic acid molecules (dsRJMAs), as well as compositions and methods for inhibiting the expression of the FLT3 gene, in particular the expression of the FLT3 gene, in a ceil tissue or mammal using such dsRNA. The invention also provides compositions and methods for treating pathological conditions and diseases caused by the expression of the FLT3 gene such as in proliferative disorders like cancer and inflammation. The invention provides double-stranded ribonucleic acid (dsRNA) molecules able to selectively and efficiently decrease the expression of FLT3, The use of FLT3 RNAi provides a method for the therapeutic and/or prophylactic treatment of diseases/disorders which are associated with inflammation and proliferative disorders, like cancers. Particular disease/disorder states include the therapeutic and/or prophylactic treatment of inflammation, like fibrosis, and proliferative disorders, like cancers, which method comprises administration of dsRNA targeting FLT3 to a human being or animal in one preferred embodiment the described dsRNA molecule is capable of inhibiting the expression of a FLT3 gene by at least 60%, preferably by at least 70 %, most preferably by at least 80 %.
In one embodiment, the invention provides double-stranded ribonucleic acid (dsRNA) molecules for inhibiting the expression of a FLT3 gene, in particular the expression of the mammalian or human FLT3 gene. The dsRNA comprises at least two sequences that are complementary to each other. The dsRNA comprises a sense strand comprising a first sequence and an antisense strand comprising a second sequence, see sequences provided in the sequence listing and also the specific dsRNA pairs in the appended table 1 and table 2. In one embodiment the sense strand comprises a sequence which has an identity of at least 90% to at least a portion of an mRNA encoding FLT3, Said sequence is located in a region of complementarity of the sense strand to the antisense strand, preferably within n cleotides 2-7 of the 5* terminus of the antisense strand, In one preferred embodiment the dsRNA specifically targets the human FLT3 gene.
In one embodiment, the antisense strand comprises a nucleotide sequence which is substantially complementary to at least part of an mRNA encoding said FLT3 gene, and the region of complementarity is most preferably less than 30 nucleotides in length. Furthermore, it is preferred that the length of the herein described Inventive dsRNA molecules (duplex length) is in the range of about 16 to 30 nucleotides, in partictdar in the range of about 18 to 28 nucleotides. Particularly useful in context of this invention are duplex lengths of about 19, 20. 21, 22, 23 or 24 nucleotides. Most preferred are duplex stretches of 19, 21 or 23 nucleotides. The dsR A, upon delivery to a cell expressing a FLT3 gene, inhibits the expression of a FLT3 gene in vitro by at least 60%, preferably by at least 70%, and most preferably by 80%, Appended Table 1 relates to preferred molecules to be used as dsRNA in accordance with this invention. Also modified dsRNA molecules are provided herein and are in particular disclosed in appended table 2, providing illustrative examples of modified dsRNA molecules of the present invention. As pointed out herein above, Table 2 provides for illustrative examples of modified dsRNAs of this invention (whereby the corresponding sense strand and antisense strand is provided in this table). The relation of the unmodified preferred molecules shown in Table 1 to the modified dsRNAs of Table 2 is illustrated in Table 5. Yet, the illustrative modifications of these constituents of the inventive dsRNAs are provided herein as examples of modifications.
Tables 3 and 4 provide for selective biological, clinical and pharmaceutical relevant parameters of certain dsRNA molecules of this invention.
Some of the preferred dsRNA molecules are provided in the appended table 1 and, inter alia and preferably, wherein the sense strand is selected from the group consisting of the nucleic acid sequences depicted in SEQ ID NOs: 3, 5, 7, 1 1, 13, 21, 25 and 2? and the antisense strand is selected from the group consisting of the nucleic acid sequences depleted in SEQ ID NOs: 4, 6, 8, 12, 14, 22, 26 and 28 Accordingly, the inventive dsRNA molecule may, Inter alia, comprise the sequence pairs selected from the group consisting of SEQ ID NOs: 3/ 45 5/6, 7/8, 1 1 12, 13/14, 25/22, 25/26 and 27/28. in the context of specific dsRNA molecules provided herein, pairs of $F,Q D NOs relate to corresponding sense and antisense strands sequences (5' to 3') as also shown in the tables. In one embodiment the dsRNA molecules comprise an antisense strand with a T overhang of 1-5 nucleotides In length, preferably 1 -2 nucleotides in length. Preferably said overhang of the antisense strand comprises uracil or nucleotides which are complementary to the mRNA encoding FLT.I. !n another preferred embodiment, said dsRNA molecules comprise a sense strand with a 3* overhang of 1-3 nucleotides in length, preferably 1-2 nucleotides in length. Preferably said overhang of the sense strand comprises uracil or nucleotides which are identical to the mRNA encoding FLT3.
In another preferred embodiment, the dsRNA molecules comprise a sense strand with a 3' overhang of 1-5 nucleotides in length, preferably 1-2 nucleotides in length, and an antisense strand with a 3* overhang of 1-5 nucleotides in length, preferably 1-2 nucleotides in length. Preferably said overhang of the sense strand comprises uracil or nucleotides which are at least 90% identical ίο the nsRNA encoding FLT3 and said overhang of the antisense strand comprises uracil or nucleotides which are at least 90% complementary to the RNA encoding FLT3.
The ds A molecules of the invention may be comprised of naturally occurring nucleotides or may be comprised of at least one modified nucleotide, such as a 2"~0~methyi 5 modified nucleotide, inverted deox thymidine, a nucleotide comprising a 5!~phosphorothioa†e group, and a terminal nucleotide linked to a choiesteryl derivative or dodecanoic acid blsdec lamide group. 2' modified nucleotides may have the additional advantage that certain immunosiimulatory factors or cytokines are suppressed when the inventive dsRNA molecules are employed in vivo, for example in a medical setting, Alternatively and non-limiting, the
10 modified nucleotide may be chosen from the group of: a 2'~deoxy-21-fltioro modified nucleotide, a 2!-deoxy~modified nucleotide, a locked nucleotide, an abasie nucleotide, 2'~arnino~modified nucleotide, 25-a!kyl~modified nucleotide, morpholino nucleotide, a phosphoramidate, and a non- natural base comprising nucleotide. In one preferred embodiment the dsRNA molecules comprises at least one of the following modified nucleotides: a 2'~Omethyl modified nucleotide,
I S a nucleotide comprising a 5'«phosphoiothioate group and a deoxythymidsne. Preferred dsRNA molecules comprising modified nucleotides are given in table 2. in another preferred embodiment one of those deoxythyrmdme nucleotides at the 3' of both strand is a inverted deoxythymi ine,
In a preferred embodiment the inventive dsRNA molecules comprise modified 0 nucleotides as detailed in the sequences given in table 2. In one preferred embodiment the inventive dsRNA molecule comprises sequence pairs selected from the group consisting of SEQ ID NOs: 3/ 4, 5/6, 7/8, 1 1/12, 13/14, 21/22, 25/26 and 27/28, and comprises overhangs at the antisense and'' or sersse strand of i~2 deoxythymidines, In one preferred embodiment the inventive dsRNA molecule comprises sequence pairs selected from the group consisting of SEQ 5 ID NOs: 3/ 4, 5/6, 7/8, 1 1/12, 13/14, 21/22, 25/26 and 27/28, and comprise modifications as detailed in table 2. Preferred dsRNA molecules comprising modified nucleotides are listed in table 2-4, with the most preferred dsRNA molecules depicted in SEQ ID Nos: 149/150, 151/152, 153/154, 157/158, 159/160, 567/168, 171/172, 173/174, 293/294, 295/29 nd 297/298.
In another embodiment, the inventive dsRNAs comprise modified nucleotides on 0 positions different from those disclosed in table 2. In one preferred embodiment two deoxythymidine nucleotides are found at the 3* of both strands of the dsRNA molecule, Preferably said deoxythymidine nucleotides form ais overhang. In one embodiment the dsRNA molecules of the Invention comprise a sense and an antisense strand wherein both strands have a half-Hie of at least 0.9 hoars, Irs one preferred embodiment the dsRNA molecules of the invention comprise a sense and an antisense strand wherein both strands have a half-life of at least 48 hours, preferably in human serum. In another embodiment the dsRNA molecules of the invention are non-lmmunestimulatory, e.g. do not stimulate IMF-alpha and TNF-alpha in vitro. In another embodiment the dsRNA molecules of the invention do stimulate INF-alpha and TNF-alpha in vitro to a very minor degree. in another embodiment, a nucleic acid sequence encoding a sense strand and / or an antisense strand comprised in the dsRNAs as defined herein are provided. The invention also provides tor ceils comprising at least one of the dsRNAs of the invention. The cell is preferably a mammalian cell, such as a human cell. Furthermore, tissues and/or non-human organisms comprising the herein defined dsRNA molecules are an embodiment of this invention, whereby said non-human organisms are particularly useful for research purposes or as research tools, for example in drug testing. Furthermore, the invention relates to a method for inhibiting the expression of a FLT3 gene, in particular a mammalian or human FLT3 gene, In a cell, tissue or organism comprising the following steps:
(a) introducing into the ceii> tissue or organism a double-stranded ribonucleic acid (dsRNA) as defined herein; and (b) maintaining said cell, tissue or organism produced In step (a) for a time sufficient to obtain degradation of the mRNA transcript of a FLT3 gene, thereby inhibiting expression of a FLT3 gene in a given cell.
The invention also relates to pharmaceutical compositions comprising the inventive dsRNAs of the invention. These pharmaceutical compositions are particularly usefui in the Inhibition of the expression of a FLT3 gene In a cei l, a tissue or an organism. The pharmaceutical composition comprising one or more of the dsRNA of the invention may also comprise (a) pharmaceutically acceptable earrier(s), diiuent(s) and/or excipient(s),
In another embodiment the invention provides methods for treating, preventing or managing inflammation and / or proliferative disorders like cancers which are associated with FLT3, said method comprising administering to a subject in need of such treatment, prevention or management a therapeutically or prophylactiealiy effective amount of ne or more of the dsRNAs of the invention. Preferably, said subject is a mammal, most preferably human patient. in one embodiment, the invention provides a method for treating a subject having a pathological condition mediated by the expression of a FLT3 gene. Such conditions comprise disorders associated with inflammation and. proliferative disorders, like cancers, as described above. In this embodiment, the dsRNA acts as a therapeutic agent for controlling the expression of a FLT3 gene, The method comprises administering a pharmaceutical composition of the invention to the patient (e.g., human), such that expression of a FLT3 gene is silenced. Because of their high specificity, the dsRNAs of the invention specifically target mRNAs of a FLT3 gene. In one preferred embodiment the described dsRNAs specifically decrease FLT3 niRMA levels and do not directly affect the expression and / or m NA levels of off-target genes in the ceil.
In one preferred embodiment the described dsRNA decrease FLT3 mRNA levels in the liver by at least 60%, preferably by at least 70%, most preferably by at least 80% in vivo. In another embodiment the described dsRNAs decrease FLT3 mRNA levels in vivo for at least 4 days. In another preferred embodiment, the dsRNAs of the invention are used for the preparation of a pharmaceutical composition for the treatment of inflammation and proliferative disorders, like cancer. Cancers to be treated with said pharmaceutical composition comprise but are not limited to leukemia and myeloproliferative diseases.
In another embodiment the invention provides vectors for inhibiting the expression of a FLT3 gene in a cell, in particular a FLT3 gene comprising a regulatory sequence operahly linked to a nucleotide sequence that encodes at least one sti'and of the dsRNA molecules of the invention.
In another embodiment, the invention provides a cell comprising a vector for inhibiting the expression of a FLT3 gene in a cell. Said vector comprises a regulatory sequence operahly linked to a nucleotide sequence that encodes at least one strand of the dsRNA molecule of the invention, Yet, it is preferred that said vector comprises, besides said regulatory sequence a sequence that encodes at least one "sense strand" of the inventive dsRNA and at least one "anti sense strand" of said dsRNA. It is also envisaged that the claimed cell comprises two or more vectors comprising, besides said regulator}-' sequences, the herein defined seqneoce(s) that encode(s) at least one strand of the dsRNA molecule of the invention. In one emhodimertt, the method comprises administering a composition comprising a dsRMA, wherein the dsKNA comprises a nucleotide sequence which is complementary to at least a part of an RNA transcript of a FLT3 gene of the mammal to be treated. As pointed out above, also vectors and cells comprising nucleic acid molecules that encode for at least one strand of the herein defined dsRNA molecules can be used as pharmaceutical compositions and may, therefore, also be employed in the herein disclosed methods of treating a subject in need of medical intervention, it is also of note that these embodiments relating to pharmaceutical compositions and to corresponding methods of treating a (human) subject also relate to approaches like gene therapy approaches. FLT3 specific dsRNA molecules as provided herein or nucleic acid molecules encoding individual strands of these inventive dsRNA molecules may also be inserted into vectors and used as gene therapy vectors for human patients. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Patent 5,328,470) or by stereotactic injection (see e.g., Chen et al (1994) Proc. Natl Acad. ScL USA 91 :3054-3057)„ The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene deliver)' vector can be produced intact from recombinant ceils, e.g.. retroviral vectors, the pharmaceutical preparation can include or»e or more ceils which produce the gene delivery system.
In another aspect of the invention, FLT3 specific dsRNA molecules thai modulate FLT3 gene expression activity are expressed from transcription units inserted into DNA or R A vectors (see, e.g.f Skiilern, A,, et al. International PCX Publication No. WO 00/221 13). These transgenes can be Introduced as a linear construct. & circular piasmid. or a viral vector, which can be Incorporated and inherited as a iransgene integrated into the host genome. The transgene can also be constructed to permit it to be inherited as an extraehromosomal piasmid (Gassmann, et ai, Proc. Natl. Acad. Sci, USA ( 1 95) 92: i 292).
The individual strands of a dsRNA can be transcribed by promoters on two separate expression vectors and co-transfected into a target cell. Alternatively each individual strand of the dsRNA can be transcribed by promoters both of which are located on the same expression piasmid. In a preferred embodiment a dsRNA is expressed as an inveited repeat joined by a linker polynucleotide sequence such that the dsRNA has a stem and loop structure.
The recombinant dsRNA expression vectors are preferably DNA p! asm ids or viral vectors. dsRNA expressing viral vectors can be constructed based on, but not limited to, adeno- associated vims (for a review, see Muzyezka, et al., Curr. Topics Micro. Immunol (1 92) 158:97-129)); adenovirus (see, for example, Berkner, et al.s BioTechmqtses (1998) 6:616), Rosenfeld et al, (1991 , Science 252:431 -434), and Rosenfeld et al. (1992), Cell 68: 143-155)); or alphavirus as well as others known in the art. Retroviruses have beers used to introduce a variety of genes into many different cell, types, including epithelial cells, in vitro and/or in vivo (see, e.g., Danes and Mulligan, Proc. Natl Acad. Sci. USA (1998) 85:6460-6464), Recombinant retroviral vectors capable of transducing and expressing genes inserted into the genome of a ceil can be produced by transacting the recombinant retroviral genome into suitable packaging cell lines such as PA317 and Psi~CRlP (Comette et al., 1991, Human Gene Therapy 2:5-10; Cone et aL, 1 84, Proc, Natl Acad. Sci. USA 81 :6349). Recombinant adenoviral vectors can be used to infect a wide variety of cells and tissues in susceptible hosts (e.g., rat, hamster, dog, and chimpanzee) (Hsu et al, 1992, J. Infectious Disease, 166:769), and also have the advaniage of not requiring mitoticalSy active cells for infection.
The promoter driving dsR A expression in either a DNA plasrnid or viral vector of the invention may be a eukaryotic R A polymerase 1 (e.g. ribosomal RNA promoter), RNA polymerase 11 (e.g. CMV early promoter or actio promoter or Ul snRNA promoter) or preferably RNA polymerase Hi promoter (e.g. U6 snRNA or 7S RNA promoter) or a prokaryotic promoter, for example the T7 promoter, provided the expression plasrnid also encodes T7 RNA polymerase required for transcription from a T7 promoter. The promoter can also direct transgene expression to the pancreas (see, e.g. the insulin regulatory sequence for pancreas (Bucchini et al, 1 86, Proc. Nail Acad. Sci. USA 83:251 Ϊ -2515)).
In addition, expression of the transgene can be precisely regulated, for example, by using an inducible regulatory sequence and expression systems such as a regulatory sequence that is sensitiv to certain physiological regulators, e.g., circulating glucose levels, or hormones (Doeherty et al, 1994, FASEB J. 8:20-24). Such inducible expression systems, suitable for the control of transgene expression in cells or in mammals include regulation by eedysone, by estrogen, progesterone, tetracycline, chemical inducers of dimerhaiion, and isopropyl-bela-Dl - thiogalaetopyranos ide (EPTG). A person skilled in the art would be able to choose the appropriate regulatory/promoter sequence based on the intended use of the d R A tr nsgene, Preferably, recombinant vectors capable of expressing dsRMA molecules are delivered as described below, and persist in target cells. Alternatively, viral vectors can be used that provide for transient expression of dsRNA molecules. Such vectors can be repeatedly administered as necessary. Once expressed, the dsRNAs bind to target RNA and modulate its function or expression, Delivery of dsRNA expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex-planted from the patient followed by emtroduction into the patient, or by any other means that allows for introduction into a desired target cell dsRNA expression DNA plasm ids are typically traosfected into target cells as a complex with cation 3C lipid carriers (e.g. Gligofeetamme) or non-eationic lipid-based carriers (e,g, Transk~TKOm). Multiple lipid transfections for dsRNA-mediated knockdowns targeting different regions of a single FLT3 gene or multiple FLT3 genes over a period of a week or more are also contemplated by the invention. Successful introduction of the vectors of the invention into host cells can be monitored using various known methods. For example, transient transfection can be signaled with a reporter, such as a fluorescent marker, such as Green Fluorescent Protein (GFP), Stable transfection of ex vivo cells can be ensured using markers that provide the transfected ceil with resistance to specific environmental factors (e.g., antibiotics and drugs), such as hygromycin B resistance.
The following detailed description discloses how to make and use the dsRNA and compositions containing dsRNA to inhibit the expression of a target FLT3 gene, as well as compositions and methods for treating diseases and disorders caused by the expression of said FLT3 gene.
DEFINITIONS
For convenience, the meaning of certain terms and phrases used in the specification, examples, and appended claims, are provided below. If there is an apparent discrepancy between the usage of a term in other parts of this specification and its definition provided in this section, the definition in this section shall prevail.
"G," "C," "A", "U" and "T" or "dT" respectively, each generally stand for a nucleotide that contains guanine, cytosine, adenine, uracil and deoxythymidine as a base, respectively. However, the term "ribonucleotide" or "nucleotide" can also refer to a modified nucleotide, as further detailed below, or a surrogate replacement moiety. Sequences comprising such replacement moieties are embodiments of the invention. As detailed below, the herein described dsRNA molecules may also comprise "overhangs", i.e. unpaired, overhanging nucleotides which are ot directly involved in the UNA double helical structure normally formed by the herein deimed pair of "sense strand" and "anti sense strand", Often, such an overhanging stretch comprises the deoxythymidine nucleotide, in most embodiments, 2 deosyihymidines in the 3' end, Such overhangs will be described and illustrated below.
The term "FLT3" as used herein relates In particular to the frns-like receptor tyrosine kinase 3 said term relates to the corresponding gene, encoded mRNA, encoded prolem/poiypeptide as well as functional fragments of the same, Preferred is the human FLT3 gene. In other preferred embodiments the ds NAs of th invention target the FLT3 gene of human (H.sapiens) and eynornoigous monkey (Macaca iascicularis) FLT3 gene. Also dsRMAs targeting the rat (Rattus norvegicus) and mouse (Miss musculus) FLT3 gene are part, of this invention. The term "FLT3 gene/sequence" does not only relate to (the) wild-type sequence(s) hut also to mutations and alterations which may be comprised in said gene/sequence. Accordingly, the present invention is not limited to the specific dsRNA molecules provided herein, The invention also reiaies to dsRNA molecules that comprise an antisense strand that is at least 85% complementary to the corresponding nucleotide stretch of a RNA transcript of a FLT3 gene that comprises such mu lons aJterations,
As used herein, "target sequence" refers to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during the transcription of a FLT3 gene, including mRNA that is a product of RNA processing of a primary transcription product.
As used herein, the term "strand comprising a sequence" refers to an oligonucleotide comprising a chain of nucleotides that is described by the sequence referred to using the standard nucleotide nomenclature. However, as detailed herein, such a "strand comprising a sequence" may also comprise modifications, like modified nucleotides. As used herein, and unless otherwise indicated, the term "complementary," when used to describe a first nucleotide sequence in relation to a second nucleotide sequence, refers to the ability of an oligonucleotide or polynucleotide comprising the first nucleotide sequence to hybridize and form a duplex structure under certain conditions with an oligonucleotide or polynucleotide comprising the second nucleotide sequence. "Complementary" sequences, as used herein, may also include, or be formed entirely from, non- Watson-Crick base pairs and/or base pairs formed from non-natural and modified nucleotides, in as tar as the above requirements with respect to their ability to hybridize are fulfilled,
Sequences referred to as "fully complementary" comprise base-pairing of the oligonucleotide or polynucleotide comprising the first nucleotide sequence to the oligonucleotide or polynucleotide comprising the second nucleotide sequence over the entire length of the first and second nucleotide sequence.
However, where a first sequence is referred to as "substantially complementary" with respect to a second sequence herein, the two sequences can be Miy complementary, or they may form one or more, but preferably not more than 13 mismatched base pairs upon hybridization, The terms "complementary", "fully complementary" and "substantially complementary" herein may be used with respect to the base matching between the sense strand and the antisense strand of a dsR A, or between the andsense strand of a dsRNA and a target sequence, as will be understood from the context of their use.
The term "double-stranded NAM, "dsRNA molecule", or "dsRNA", as used herein, refers to a ribonucleic acid molecule, or complex of ribonucleic acid molecules, having a duplex structure comprising two anti-parallel and substantially complementary nucleic acid strands. The iwo strands forming the duple structure may he different portions of one larger RNA molecule, or they may be separate RNA molecules. Where the two strands are part of one larger molecule, and therefore are connected by an uninterrupted chain of nucleotides between the 3 '-end of one strand and the 5' end of the respective other strand forming the duplex structure, the connecting RNA chain is referred to as a "hairpin loop". Where the two strands are connected covaiently by means other than an uninterrupted chain of nucleotides between the 3 '-end of one strand and the 5*end of the respective other strand forming the duplex structure, the connecting structure is referred to as a "linker". The RNA strands may have the same or a different number of nucleotides. In addition to the duplex structure, a dsRNA may comprise one or more nucleotide overhangs. The nucleotides in said "overhangs" may comprise between 0 and 5 nucleotides, whereby "0s* means no additional nucieotide(s) that form(s) an "overhang" and whereas ;i5" means ftve additional nucleotides on the individual strands of the dsRNA duplex. These optional "overhangs" are located in the 35 end of the individual strands. As will be detailed below, also dsRNA molecules which comprise only an "overhang" in one of the two strands may be useful and even advantageous in context of this invention, The "overhang" comprises preferably between 0 and 2 nucleotides. Most preferably 2 "dT" (deoxythymldine) nucleotides are found at the V end of both strands of the dsRMA, Also 2 "U5'(uraeil} nucleotides cars be used as overhangs at the 3' end of both strands of the dsRNA. Accordingly, a "nucleotide overhang" refers to the unpaired nucleotide or nucleotides that protrude from the duplex structure of a dsRNA when a 3'~end of one strand of the dsRNA extends beyond the 5'-end of the other strand, or vice versa. For example the antisense strand comprises 23 nucleotides and the sense strand comprises 21 nucleotides, forming a 2 nucleotide overhang at the 3' end of the antisense strand. Preferably, the 2 nucleotide overhang is fully complementary to the m NA of the target gene. "Blunt" or "blunt end" means that there are no unpaired nucleotides at that end of the dsRNA, i.e., no nucleotide overhang, A "blunt ended" dsR A is a dsRNA that is double-stranded over its entire length, i.e., no nucleotide overhang at either end of the molecule.
The term "antisense strand" refers to the strand of a dsRNA which includes a region that Is substantially complementary to a target sequence. As used herein, the term "region of complementarity" refers to the region on the antisense strand, that is substantially complementary to a sequence, for example a target sequence. Where the region of complementarity is not fully complementary to the target sequence, the mismatches are most tolerated outside nucleotides 2-7 of the 5' terminus of the antisense strand
The term "sense strand," as used herein, refers to the strand of a dsRNA that includes a region that is substantially complementary to a region of the antisense strand. "Substantially complementary" means preferably at least S5% of the overlapping nucleotides in sense and antisense strand are complementary.
"Introducing into a cell", when referring to a dsRNA, means tacilit ing uptake or absorption into the cell, as is understood by those skilled in the art. Absorption or uptake of dsRNA can occur through unaided diffusive or active cellular processes, or by auxiliary agents or devices, The meaning of this term is not limited to cells in vitro; a dsRNA may also be "introduced into a cell", wherein the ceil is part of a living organism. In such instance, introduction into the cell will include the delivery to the organism. For example, for in vivo delivery, ds&NA can be injected Into a tissue site or administered systemically, it is, for example envisaged that the dsRNA molecules of this invention be administered to a subject in need of medical, intervention. Such an administration may comprise the injection of the dsRNA, the vector or a cell of this invention into a diseased site in said subject, for example into liver tissue/cells or into cancerous tissues cells, like liver cancer tissue. In addition, the injection is preferably in close proximity to the diseased tissue envisaged. In vitro introduction into a ceil Includes methods known in the art such as electroporation and lipofeeiion,
As used, herein, "proliferating" and "proliferation" refer to cells undergoing mitosis. Throughout this application, the term "proliferative disorder" refers to any disease/disorder marked by unwanted or aberrant proliferation of tissue. As used herein, the term " proliferative disorder" also refers to conditions in which the unregulated and/or abnormal growth of cells can lead to the development of an unwanted condition or disease, which can be cancerous or noncancerous.
The term "inflammation" as used herein refers to the biologic response of body tissue to injury, irritation, or disease which can be caused by harmful stimuli, for example, pathogens, damaged cells, or irritants. Inflammation is typically characterized by pain and swelling. Inflammation is intended to encompass both acute responses, in which inflammatory processes are active (e.g., neutrophils and leukocytes), and chronic responses, which are marked by slow- progress, a shift in the type of cell present at the site of inflammation, and the formation of connective tissue. One example of an inflammation-caused disease is fibrosis.
Cancers to be treated comprise, but are again not limited to leukemia, AML, solid tumors, liver cancer, brain cancer, breast cancer, lung cancer, NSCLC, colorectal cancer, bladder cancer and prostate cancer, whereby said liver cancer may, inter alia, be selected from the group consisting of hepatocellular carcinoma (HCC), hepatoblastoma, a mixed liver cancer, a cancer derived from mesenchymal tissue, a liver sarcoma or a cholangiocarcinoma.
The terms "silence**, "inhibit the expression of and "knock down" in as far as they refer to a FLT3 gene, herein refer to the at least partial suppression of the expression of a FLT3 gene, as manifested by a reduction of the amount of niRNA transcribed from a FLT3 gene which may be isolated from a first cell or group of cells in which a FLT3 gene is transcribed and which has or have been treated such that the expression of a FLT3 gene is inhibited, as compared to a second cell or group of cells substantially identical to the first cell or group of ceils but which has or have not been so treated (control cells). The degree of inhibition is usually expressed in terras of
{mRNA in control cells) - {mRNA in treated cells) . ΛΛ.„ .
un 'NA in control cells) Alternative y, the degree of inhibition may be given in terms of a reduction of a parameter that is functionally linked to the FLT3 gene transcription, e.g. the amount of protein encoded by a FLT3 gene which s secreted by a cell, or the number of cells displaying a certain pherrotype. As illustrated in the appended examples and in the appended tables provided herein., the inventive dsRNA molecules are capable of inhibiting the expression of a human FLT3 by at least about 60%, preferably by at least 70%, most preferably by at least S0% in vitro assays, Le in vitro. The term "in vitro" as used herein includes but is not limited to cell culture assays. In another embodiment the inventive dsRNA molecules are capable of inhibiting the expression of a mouse or rat FLT3 by at least 60 %.preferably by at least 70%, most preferably by at least 80%. The person skilled in the art can readily determine such an inhibition rate and related effects, in particular in light of the assays provided herein.
The term "off target" as used herein refers to all non-target mRKAs of the transcriptorne that are predicted by in silieo methods to hybridize to the described ds'R As based on sequence complementarity. The dsRNAs of the present invention preferably do specifically inhibit the expression of FLO, i.e. do not Inhibit the expression of any off-target
The term "half-Site" as used herein is a measure of stability of a compound or molecule and can be assessed by methods known to a person skilled in the art, especially in light of the assays provided herein. The term "non-imrnunost!mulatory'' as used herein refers to the absence of any induction of a immune response by the invented dsRNA molecules. Methods to determine immune responses are well known to a person skilled in the art for example by assessing the release of cytokines, as described in the examples section,
The terms "treat", "treatment", and the like, mean in context of this invention the relief from or aiieviation of a disorder related to FLT3 expression, like inflammation and proliferative disorders, like cancers.
As used herein, a "pharmaceutical composition" comprises a pharmacologically effective amount of a dsRNA and a pharmaceutically acceptable carrier. However, such a "pharmaceutical composition" may also comprise individual strands of such a dsRNA molecule or the herein described veetoris) comprising a regulatory sequence operabiy linked to a nucleotide sequence thai encodes at least one strand of a sense or an antisense strand comprised In the dsRNAs of this invention. It is also envisaged that cells, tissues or isolated organs that express or comprise the herein defined dsRNAs may be used as "pharmaceutical compositions". As used herein, "pharmacologically effective amount," '^therapeutically effective amount" or simply "effective amount" refers to thai amount of an RNA effective to produce the intended pharmacological, therapeutic or preventive result.
The term "pharmaceutically acceptable carrier" refers to a carrier for admin istration of a therapeutic agent Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, etbano!, and combinations thereof. The term specifically excludes cell culture medium. For drugs administered orally, pharmaceutically acceptable carriers include, but are not limited to pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives as known to persons skilled in the art.
It is in particular envisaged that the pharmaceutically acceptable carrier allows for the systemic administration of the dsRNAs, vectors or cells of this invention. Whereas also the enteric administration is envisaged the parenteral administration and also transdermal or transmueosaS (e.g. insufflation, buccal, vaginal, anal) administration as well as inhalation of the drug are feasible ways of administering to a patient in need of medical intervention the compounds of this invention. When parenteral administration is employed, this can comprise the direct injection of the compounds of this invention into the diseased tissue or at least in close proximity. However, also intravenous, intraarterial subcutaneous, intramuscular, intraperitoneal, intradermal intrathecal and other administrations of the compounds of this invention are within the skill of the artisan, for example the attending physician.
For intramuscular, subcutaneous and intravenous use, the pharmaceutical compositions of the invention will generally be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pli and Isotonicity. In a preferred embodiment, the carrier consists exclusively of an aqueous buffer. In this context, "exclusively" means no auxiliary agents or encapsulating substances are present which might affect or mediate uptake of dsRNA In the cells that express a FLT3 gene. Aqueous suspensions according to the invention may include suspending agents such as cellulose derivatives, sodium alginate, poly vinyl-pyrrol idone and gum tragacanth, and a wetting agent such as lecithin. Suitable preservatives for aqueous suspensions include ethyl and n~prop i p-hydrox benzoate. The pharmaceutical compositions useful according to the invention also include encapsulated formulations to protect the dsRNA against rapid elimination from the body, such as a controlled release formulation, including Implants and microencapsulated deliver)' systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and potylactic acid, Methods for preparation of such formulations will be apparent to those skilled in the art. Liposomal suspensions can also be used as pharmaceutically acceptable carriers. These ears be prepared according to methods known to those skilled in the art, for example, as described in PCX publication WO 91/06309 which is incorporated by reference herein.
As used herein, a "transformed cell" is a cell Into which at least one vector has been Introduced from which a dsHNA molecule or at least one strand of such a dsRNA molecule may be expressed. Such a vector is preferably a vector comprising a regulatory sequence operabiy linked to nucleotide sequence that encodes at least one sense strand or antisense strand of a dsRNA of the present invention.
It can be reasonably expected that shorter dsRNAs comprising one of the sequences in Table 1 and 4 minus only a few nucleotides on one or both ends may be similarly effective as compared to the dsRNAs described above.
In one preferred embodiment the inventive dsRNA molecules comprise nucleotides 1-1 of the sequences given in Table 1.
As pointed out above, in most embodiments of this invention, the ds MA molecules provided heresn comprise a duplex length (i.e. without "overhangs") of about 16 to about 30 nucleotides, Particular useful dsRNA duplex lengths are about 19 to about 25 nucleotides. Most preferred are duplex structures with a length of 19 nucleotides, in the inventive dsRNA molecules, the antisense strand is at least partially complementary to the sense strand.
The dsRNA of the invention can contain one or more mismatches to the target sequence. In & preferred embodiment, the dsRNA of the invention contains no more than 13 mismatches, if the antisense strand of the dsRNA contains mismatches to a target sequence, it is preferable that the area of mismatch not be located within nucleotides 2-7 of the 5' terminus of the antisense strand. In another embodiment it is preferable that the area of mismatch not be located within nucleotides 2-9 of the 5' terminus of the antisense strand. As mentioned above, at least one end/strand of the ds NA may have a single-stranded nucleotide overhang of 1 to 5, preferably I or 2 nucleotides, ds NAs having at least one nucleotide overhang have unexpectedly superior inhibitory properties than their blunt-ended counterparts. Moreover, the present inventors have discovered that the presence of only one nucleotide overhang strengthens the interference activity of the dsRNA, without affecting its overall stability. dsRNA having only one overhang has proven particularly stable and effective in vivo, as well as in a variety of cells, cell culture mediums, blood, and serum. Preferably, the single-stranded overhang is located at the S'-terminal end of the antisense strand or, alternatively, at the S'-term al end of the sense strand. The dsRNA may also have a blunt end, preferably located at the S'-end of the antisense strand. Preferably, the antisense strand of the dsRNA has a nucleotide overhang at the 3?-end, and the 5 '-end is blunt. In another embodiment, one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate.
The dsRNA of the present invention may also be chemically modified to enhance stability. The nucleic acids of the invention may be synthesized and/or modified by methods well established in the art, such as those described in "Current protocols in nucleic acid chemistry", Beaueage, S.L. et al. (Edrs.), John Wiley & Sons. Inc., New York, NY, USA, which is hereby incorporated herein by reference. Chemical modifications may include, but are not limited to 2' modifications, introduction of non-natural bases, eovalent attachment to a ligand, and replacement of phosphate linkages with thiophosphate linkages, inverted deoxythyniidines, in this embodiment, the integrity of the duplex structure is strengthened by at least one, and preferably two, chemical linkages. Chemical linking may be achieved by any of a variety of well-known techniques, for example by introducing eovalent, ionic or hydrogen bonds; hydrophobic interactions, van der Waais or stacking interactions; by means of metal-ion coordination, or through use of purine analogues, Preferably, the chemical groups that can be used to modify the dsRNA include, without limitation, methylene blue; blfonctional groups, preferably bis-(2-ehloroethyl)annne; N-acetyi-N'^p-glyoxylbenzoyiJcystamine 4-thiouracii; and psoralen, in one preferred embodiment, the linker is a hexa-ethylene glycol linker. n this case, the dsRNA are produced by solid phase synthesis and the hexa-eihylene glycol linker is incorporated according to standard methods (e.g., Williams, DX, and ,B. Hall, Bioch m. (1996) 35: 14665-14670), In a particular embodiment, the S'-ersd of the antisense strand and the 3'-end of the sense strand are chemically linked via a hexaethylene glycol linker, in another embodiment, at least one nucleotide of the dsRNA comprises a phosphorothioate or phosphorodlthioaie groups. The chemical bond at the ends of the dsRNA is preferably formed by triple-helk bonds,
In certain embodiments, a chemical bond may be formed by means of one or several bonding groups, wherein such bonding groups are preferably poiy-(oxyphosphinicooxy-l ;3- propandiol)- and/or polyethylene glycol chains. In other embodiments, a chemical bond may also be formed by means of purine analogs introduced into the double-stranded structure instead of purines. in further em odiment, a chemical bond may be formed by azahen^ene units introduced into the double-stranded structure. In still further embodiments, a chemlcai bond may be formed by branched nucleotide analogs instead of nucleotides introduced into the double- stranded structure. In certain embodiments, a chemical bond may be induced by ultraviolet light.
In yet another embodiment, the nucleotides at one or both of the two single strands may be modified to prevent or Inhibit the activation of cellular enzymes, for example certain nucleases. Techniques for inhibiting the activation of cellular enzymes are known in the art including, but not limited to, 2' -amino modifications, 2?-amino sugar modifications, 2'-F sugar modifications, 2'-F modifications, 2'~alkyl sugar modifications, uncharged backbone modifications, morpholino modifications, 2'~Q~methy! modifications, and phosphoramidate (see, e.g., Wagner, Nat Med, (1995) 1 : 1 1 16-8). Thus, at least one 2 '-hydroxy! group of the nucleotides on a dsRMA is replaced by a chemical group, preferably by a 2*-amino or a 2'- rnethyl group. Also, at least one nucleotide may be modified to form a locked nucleotide. Such locked nucleotide contains a methylene bridge that connects the 2* -oxygen of ribose with the 4s- carbon of ribose. Introduction of a locked nucleotide into an oligonucleotide improves the affinity for complementary sequences and increases the melting temperature by several degrees.
Modifications of dsRNA molecules provided herein may positively influence their stability in vivo as well as in vitro and also improve their delivery to the (diseased) target side, Furthermore, such structural and chemical modifications may positively influence physiological reactions towards the dsRNA molecules upon administration, e.g. the cytokine release which is preferably suppressed. Such chemical and structural modifications are known in the ait and are, inter alia, illustrated in Nawrot (2006) Current Topics in Med Chem, 6, 13-925.
Conjugating a iigand to a dsRNA can enhance its cellular absorption as well as targeting lo a particular tissue, in certain instances, a hydrophobic ligand is conjugated to the dsRNA to facilitate direct permeation of the cellular membrane. Alternatively, the ligand conjugated to the ds NA is a substrate for receptor-mediated endocytosis. These approaches have been used to facilitate cell permeation of antisense oligonucleotides. For example, cholesterol has been conjugated to various antisense oligonucleotides resulting in compounds that are substantially more active compared to their non-conjugated analogs. See M. Manoharan Antisense & Nucleic Acid Drug Development 2002, 12t 103. Other lipophilic compounds that have been conjugated to oligonucleotides include 1-pyrene butyric acid, t3-his-0-(hexadecyi)glyeerol, and menthol. One example of a Hgand for receptor-mediated endocytosis is folic acid. Folic acid enters the cell by toiate-receptor-mediated endocytosis. dsRNA compounds bearing folic acid would be efficiently transported into the cell via the folate-receptor-rnediated endocytosis. Attachment of folic acid to the 3 '-terminus of an oligonucleotide results in increased cellular uptake of the oligonucleotide (Li, S.; Deshniukh, H. M„; Huang, L. Ph rm. Res, 1998, 15, 1540). Other ligands that have been conjugated to oligonucleotides include polyethylene glycols, carbohydrate clusters, cross-linking agents, porphyrin conjugates, and delivery peptides. in certain instances, conjugation of a cationic Hgand to oligonucleotides often results in improved resistance to nucleases. Representative examples of cationic ligands are propylammonium and dimethylpropylairimonium. Interestingly, antisense oligonucleotides were reported to retain their high binding affinity to mRNA when the cationic hgand was dispersed throughout the oligonucleotide. See ML Manoharan Antisense & Nucleic Acid Drug Development 2002, 12, 103 and references therein, The ligand-eonjugated dsRNA of the invention may be synthesized hy the use of a dsR A that bears a pendant reactive functionality, such as that derived from the attachment of a linking molecule onto the dsRMA. This reactive oligonucleotide may be reacted directly with commercially-available ligands, ligands that are synthesized bearing any of a variety of protecting groups, or ligands that have a linking moiety attached thereto. The methods of the invention facilitate the synthesis of ligand-eonjugated dsRNA hy the use of, in some preferred embodiments, nucleoside monomers that have been appropriately conjugated with ligands and that may further be attached to a solid-support material. Such !igand-nucleoside conjugates, optionally attached to a solid-support material, are prepared according to some preferred embodiments of the methods of the invention via reaction of a selected serum-binding ligand with a linking moiety located on the 5' position of a nucleoside or oligonucleotide, in certain instances, an dsRNA bearing an aralkyl ligand attached to the S'-terminus of the dsRNA is prepared by first covalently attaching a monomer building block to a control ied-pore-glass support via a long-chain arninoaikyi group. Then, nucleotides are bonded via standard solid- phase synthesis techniques to the monomer building-block bound to tJhe solid support. Hie monomer building block may be a nucleoside or other organic compound thai is compatible with solid-phase synthesis. The dsRNA used ire the conjugates of the invention may be conveniently and routinely made through the well-known technique of solid-phase synthesis. It is also known to use similar techniques to prepare other oligonucleotides, such as the phosphorothioates and alkylated derivatives.
Teachings regarding the synthesis of particular modified oligonucleotides may he found in the following U.S. patents: U.S. Pat, No, 5,218,105, drawn to polyamine conjugated oligonucleotides; U.S. P&L Nos. 5,541,307, drawn to oligonucleotides having modified backbones; U.S. Fat. No. 5,521,302, drawn to processes for preparing oligonucleotides having chiral phosphorus linkages; U.S. Pat No, 5,539,082, drawn to peptide nucleic acids; U.S. Pat. No. 3,554,746, drawn to oligonucleotides having β-laetam backbones; U.S. Pat. No. 5,571,902, drawn to methods and materials for the synthesis of oligonucleotides; U.S. Pat, No. 5,578,718, drawn to nucleosides having alkyithio groups, wherein such groups may be used as linkers to other moieties attached at any of a variety of positions of the nucleoside; U.S. Pat, No 5,587,361 drawn to oligonucleotides having phosphorothioate linkages of high ehlral purity; U.S. Pat, No. 5,506,351, drawn to processes for the preparation of 2 -O-a!kyI guanosine and related compounds, including 2,6~diaminopurine compounds; U.S. Pat. No. 5,587,469, drawn to oligonucleotides having N-2 substituted purines; U.S. Fat. No. 5,587,470, drawn to oligonucleotides having 3-dea apurines; U.S. Pat, No. 5,608,046, hoth drawn to conjugated 4'- desrnethyl nucleoside analogs; U.S. Pat. No. 5,610,289, drawn to backbone-modified oligonucleotide analogs; U.S. Pat, No 6,262,241 drawn to, inter alia, methods of synthesizing 2'- flitoro-oligoui!cleotides.
In the Hgand-conjugated dsRNA and ligand-molecuie bearing sequence-specific linked nucleosides of the invention, the oligonucleotides and oligonucieosides may be assembled on a suitable DNA synthesizer utilizing standard nucleotide or nucleoside precursors, or nucleotide or nucleoside conjugate precursors that already bear the linking moiety, ligand-nucleoiide or nucleoside-eonjugate precursors that already hear the ligand molecule, or non-nacleoslde ligaitd- bearing building blocks. When using nudeotide-eonjugate precursors thai already bear a linking moiety, the synthesis of the sequence-specific linked nucleosides is typically completed, and the ligand molecule Is then reacted with the linking moiety to form the ligand-conjugated oligonucleotide. Oligonucleotide conjugates bearing a variety of molecules such as steroids, vitamins, lipids and reporter molecules, has previously been described (see Manoharan et al, PCX Application WO 93/07883). In a preferred embodiment, the oligonucleotides or linked nucleosides of the invention are synthesized by an automated synthesizer using phosphoramidites derived from lijgand~nudeos.de conjugates in addition to commercially available phosphoramidites.
The incorporation of a 2f~0~methyl, 2*-Q-eihyl, 2'-0-propyl. 2'-0-allyl, 2'-0-aminoalkyl or 2!-deoxy-2'-fluoro group in nucleosides of an oligonucleotide confers enhanced hybridization properties to the oligonucleotide. Further, oligonucleotides containing phosphorothtoate backbones have enhanced nuclease stability. Thus, functionalized, linked nucleosides of the invention can be augmented to include either or both a phosphorothioate backbone or a 2 -0- methyl, 2'-0-ethyi, 2"~0~propyl, 2i-0-aminoaikyiJ 2"-0-allyl or 2'-deGxy-2'~i!uGro group. In some preferred embodiments, functional ized nucleoside sequences of the invention possessing an amino group at the S'-terminus are prepared using a DNA synthesizer, and then reacted with an active ester derivative of a selected ligand. Active ester derivatives are well known to those skilled in the art. Representative active esters include N-hydrosuccinimide esters, tetrafluorophenolic esters, pentaflnorophenolic esters and pentachlorophenoiic esters. The reaction of the amino group and the active ester produces an oligonucleotide in which the selected ligand is attached to the S'-position ihrough a linking group. The amino group at the 5'~ terminus can be prepared utilizing a 5'-Am mo-Modifier C6 reagent. In a preferred embodiment, ligand molecules may be conjugated to oligonucleotides at the 5 -position by the use of a ligand- nucleoside phosphoramidite wherein the ligand is linked to the 5'-hydroxy group directly or indirectly via a linker. Such ligand-nucleoside phosphoramidites are typically used at the end of an automated synthesis procedure to provide a Hgand-conjugated oligonucleotide bearing the ligand at the 5'~terminus.
In one preferred embodiment of the methods of the invention, the preparation of ligand conjugated oligonucleotides commences with the selection of appropriate precursor molecules upon which to construct the ligand molecule. Typically, die precursor is an appropriately- protected derivative of the eommonSy-used nucleosides. For example, the synthetic precursors for the synthesis of the ligand-conjugated oligonucleotides of the invention include, but are not limited to, 2'-ammoaikoxy~5t-ODMT-n«cSeosidess, 2,-6-aniinoaikyla ino-5'-GDMT-nueleosides, S'-S-aminoaikoxy-^ -deoxy-nueleosides, 5'-6~aminoaIkoxy-2-proteeted-niscieosides, 3 -6- amsnoalkoxy-5'-ODMT-nucleosides, and S'-aminoalkyiamiriO-S'-ODMT-rsucIeosidos that may be protected in the nucleohase portion of the molecule. Methods for the synthesis of such amino- linked protected nucleoside precursors are know to those of ordinary skill in the art.
In many cases, protecting groups are used during the preparation of the compounds of the invention. As used herein, the term "protected" means that the indicated moiety has a protecting group appended thereon. In some preferred embodiments of the invention, compounds contain one or more protecting groups. A wide variety of protecting groups can be employed in the methods of the invention, in general, protecting groups render chemicai functionalities inert to specific reaction conditions, and can be appended to and removed from such functionalities in a molecule without substantially damaging the remainder of the molecule.
Representative hydroxy! protecting groups, as well as other representative protecting groups, are disclosed in Greene and Wu s, Protective Groups m Organic Symhesis, Chapter 2, 2d ed,, John Wiley 8c Sons, New York, 1991, and Oligonucleotides And Analogues A Practical Approach, Ekstein, F. Ed., IRL Press, M.Y, 1 91 ,
Amino-protecting groups stable to acid treatment are selectively removed with base treatment and are used to make reactive amino groups selectively available for substitution. Examples of such groups are the Fmoc (E. Atherton and R. C. Sheppard in The Peptides, S. Udenfriersd, J. Meienhofer, Eds.s Academic Press, Orlando, 1987, volume 9, p,l) and various substituted sulfonyiethy! carbamates exemplified by the Nsc group (Samukov et al. Tetrahedron LetL, 1994, 35:7821.
Additional amino-protecting groups include, but are not limited to, carbamate protecting groups, such as 2 rhnethylsilylethoxycarbon I (Teoc), 1 -methyl- 1 -{4- biphenylyi)ethoxycarbonyS (Bpoc), t-butoxycarbonyi (BOG), allyloxycarbonyS (Alloc), 9~ fluorenylmethyloxyearbonyl (Fmoc), and benzy!oxycarbonyl (Cbz); amide protecting groups, such as formyl, acetyl trihaloaeetyl, benzoyl, and mtrophenylaeetyS; sulfonamide protecting groups, such as 2-nitrobenzenesu!fonyl; and imine and cyclic imide protecting groups, such as phthalimido and diihiasuecmoyt Equivalents of these amino-protecting groups are also encompassed by the compounds and methods of the invention, Many solid supports are commercially available and one of ordinary skill in the art can readily select a solid support to be used in the solid-phase synthesis steps, in certain embodiments, a universal support is used. A universal support allows for the preparation of oligonucleotides having unusual or modified nucleotides located at the 35 erminus of the oligonucleotide. For further details about universal supports see Scott et ah, JnttovaHom and Perspectives in solid-phase Synthesis, 3rd International Symposium, 1.994, Ed. Roger Epton, Mayflower Worldwide, 115-124]. In addition, it has been reported that the oligonucleotide cars be cleaved from the universal support under milder reaction conditions when the oligonucleotide is bonded to the solid support via a i>'»-lt2~ac £oxyphosphate group which more readily undergoes basic hydrolysis. See Guzaev, A. L; Manoharan, M, J. Am, Ch rn. Soc. 2003, 125, 2380.
The nucleosides are linked by phosphorus-containing or non-phosphorus-eontaining covaleut intemueleoside linkages, For the purposes of identification, such conjugated nucleosides can be characterized as ligand-bearing nucleosides or ligand-nucieoside conjugates. The linked nucleosides having an aralkyl ligand conjugated to a nucleoside within their sequence will demonstrate enhanced dsRNA activity when compared to like ds NA compounds that are not conjugated.
The araikyS-iigand-eonjugated oligonucleotides of the invention also include conjugates of oligonucleotides and linked nucleosides wherein the ligand is attached directly to the nucleoside or nucleotide without the intermediacy of a linker group. The ligand may preferably he attached, via linking groups., at a carboxyl, amino or oxo group of the ligand. Typical linking groups may be ester, amide or carbamate groups.
Specific examples of preferred modified oligonucleotides envisioned for use in the ligand-conjugated oligonucleotides of the invention include oligonucleotides containing modified backbones or non-natural internucieoside linkages. As defined here, oligonucleotides having modified backbones or intemueleoside linkages include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone, For the purposes of the invention, modified oligonucleotides that do not have a phosphorus atom in their intersugar backbone can also be considered to be oligonucieosides, Specific oligonucleotide chemical modifications are described below. It is not necessary for all positions in a given compound to be uniformly modified. Conversely, more than one modifications may be incorporated in a single dsRNA compound or even in a single nucleotide thereof.
Preferred modified intemucleoside linkages or backbones include, for example, phosphorothioates, ehiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalk iphosphotriesters, methyl and other alky! phosphonates including 3'-a!k lene phospSionates and chiral phosphonates, phosphinates, phosphoramidates including 3'~ammo phosphoraniidate and ammoalkylphosphoramidates, thionophosphoramidates, thionoaikyiphosphonaies, thionoalkylphosphotriesters, and boranophosp ates having normal 3!- 51 linkages, 2I~51 linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to 5?~3' or 2!~5' to 5 -2', Various salts, mixed salts and free-acid forms are also included.
Representative United States Patents relating to the preparation of the above phosphorus- atom-containing linkages include, but are not limited to, L S. Pat. Nos. 4,469,863; 5,023,243; 5,264,423; 5,321 , 131 ; 5,399,676; 5,405,939; 5,453,496; 5,455,233 and 5,466,677, each of which is herein incorporated hy reference.
Preferred modified intemucleoside linkages or backbones that do not include a phosphorus atom therein (i.e., oiigonucleosides) have backbones that are formed by short chain alkyl or cycioalkyl intersugar linkages, mixed heteroatom and aikyl or cycioalkyl iniersugar linkages, or one or more short chain heteroatomk or heterocyclic intersugar linkages. These include those having morphoHrro linkages {formed in part from the sugar portion of a nucleoside); slloxane backbones; sulfide, sulfoxide and sulfone backbones; formaeety! and ihioformacetyl backbones; methylene formacetyl and thioformacetyi backbones; alkene containing backbones; suifamate backbones; methylene imino and methyienehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts.
Representative United States patents relating to the preparation of the above oiigonucleosides include, but are not limited to, U.S. Pat Nos. 5,034,506; 5,214,134; 5,216, 14! ; 5,264,562; 5,466,677; 5,470,967; 5,489,677; 5,602,240 and 5,663312, each of which is herein incorporated by reference. In other preferred oligonucleotide mimetics, both the sugar and the internucleoslde linkage, i,e,, the backbone, of the nucleoside units are replaced with novel groups. The nucleobase units are maintained for hybridization with an appropriate nucleic acid target compound. One such oligonucleotide, an oligonucleotide mimetic, that has been shown to have excellent hybridization properiies} Is referred to as a peptide nucleic acid (PNA). In PMA compounds, the sugar-backbone of an oligonucleotide Is replaced with an amide-containing backbone, in particular an aminoethylglycine backbone. The nucieobases are retained and are bound directly or indirectly to atoms of the amide portion of the backbone. Teaching of PNA compounds can be found for example in U.S. Pat No. 5,539,082. Some preferred embodiments of the invention employ oligonucleotides with phosphorothioate linkages and oligonucleotides with heteroatorn backbones, and in particular— CH2--NH--O-CH2 --, ~CH2™N(CIl3)™0-C!I2 - [known as a methylene (methyli ino) or MM! backbone], -CH2-0-N(CH3)~CH2 ~CH2-N(CH3)- (CH3)~CH2-, and -0™NiC¾)™CH2 --C¾— [wherein the native phosphodiester backbone is represented as ~θ— p— 0— CF¾~~ ] of the above referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above referenced LIS. Pat. No. 5,602,240. Also preferred are oligonucleotides having morpholino backbone structures of the above-referenced U.S. Pat. No, 5S034S506.
The oligonucleotides employed in the ligand-conjugated oligonucleotides of the invention may additionally or alternatively comprise nucleobase (often referred to in the art simply as "base") modifications or substitutions. As used herein, "unmodified" or "natural" nucieobases include the purine bases adenine (A) and guanine (G), and the pyrlmidine bases thymine (T), cytosine (C), and uracil (U). Modified nucieobases include other synthetic and natural nucieobases, such as 5~methy!eyiosine (5~nie~C)s 5-hydroxymeihyi cytosine, xanthine, hypoxanthine, 2~aminoadenine, 6~methyl and other aikyl derivatives of adenine and guanine, 2- propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2- thiocytossne, 5-halouracil and cytosine, 5-propynyI uracil arid cytosine, 6-azo uracil, cytosine and thymine, 5-uracsI (pseudouracii), 4-thiouracil, 8-haio, 8-aniinos 8-thiol, 8-ihioaikyl, 8- hydroxyl and other 8~substituted adenines and guanines, 5-haio particularly 5-bromo, 5- trifluoromethyi and other 5-substituted uracils and cytosines, 7-methylguanine and 7- nieihyladenine, S-azaguanine and S-azaadenine, 7- ieazaguanine and 7-deazaadenine and 3- deazaguanine and 3-dsa aadenine. Further nucleobases include those disclosed in U.S. Pat, No. 3,687,808, those disclosed in the Concise Encyclopedia Of Poiymer Science And Engineering, pages §58-859, Kroschwite, J. L, ed, John Wiley & Sons, 1990, those disclosed by Engiisch et al.s Ange a ite Chemie. Internationa! Edition, 1991 , 30, 613, and tbose disclosed by Sanghvi, Y, S„, Chapter 15, A isense Research an Applications, pages 289-302, Crooke, S. T, and Lebleu, B.„ ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligonucleotides of the invention. These include 5-sufastituted pyrimidines, 6- azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-antinopropyladenine, 5- propynyluracil and S-propynyicytosine. S-Methyleytosme substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2 °C. (Id., pages 276-278) and are presently preferred base substitutions, even more particularly when combined with 2 -methoxyethyt sugar modifications.
Representative United States patents relating to the preparation of certain of the above- noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Pat. No. 3,687,808, as well as U.S. Pat. Nos 5, 134,066; 5,459,255; 5,552,540: 5,594,121 and 5,596,091 all of which are hereby incorporated by reference. in certain embodiments, the oligonucleotides employed in the Hgand-eonjugated oligonucleotides of the invention may additionally or alternatively comprise one or more substituted sugar moieties. Preferred oligonucleotides comprise one of the following at the 2" position: OH: F; 0~, S-, or N-alkyl, G-, S~, or N-alkenyl, or O, S~ or M~aikynyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted Cj to Cio alky! or C to Cjo alkeny! and alkynyl. Particularly preferred are 0[(CH2)nO]ms 0(CH2)nOCH¾ G(CH2}n H2l Ο(0¾),,ΰ¾, 0(CH2)r.O H2, and 0{€Η2)„Ο [{ΟΗ2)η€Ή3)]2. where n and ni are from 1 to about 10. Other preferred oligonucleotides comprise one of the following at the 2* position: Q to Cjo lower alkyl, substituted lower alkyl, alkaryi, aralkyt 0~alkaryi or O-araiky!, SH, SCH3, OCN, CI, Br, C , CF3> OCF3) SOCH3, S02 CH3, ON02, NO¾ N3> NH2} heterocydoalkyl heterocycloalkary!, am oalkylamino, pofyaikylamino, substituted silyi, an NA cleaving group, a reporter group, an Inierealator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving die pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties. A preferred modification includes 2 -methoxyethoxy
Figure imgf000027_0001
also known as 2'-G-(2~methoxyethyi) or 2'- MOE], i.e., an alkoxyalkoxy group. A further preferred modification includes 2'- dimethylaminooxyethoxy, i.e., a 0{€¾)2θΝ{ί¾)2 group, also known as 2'-DMAOE, as described in U.S. Pat. No. 6, 127,533, filed on Jan. 30, 1998, the contents of which are incorporated by reference.
Other preferred modifications include 2'-methoxy (2!-0~Cl¾), 2'-aminopropoxy (2!- OCH2CH2CH2NH2) and 2 -fluoro (2'~F), Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3' osition of the sugar on the 3' terminal nucleotide or in 2'~5' linked oligonucleotides.
As used herein, the term "sugar substituent group" or "2'-subsi!tueni group" includes groups attached to the mposition of the nbofiiranosyl moiety with or without an oxygen atom, Sugar substituent groups include, but are not limited to, fluoro, Q-alky!, O-alkylamino, O- alkylaikoxy, protected O-alkylamino, O-afkylar noalkyi, Oaiky! imidazole and polyethers of the formula (0-alkyl)ro, wherein rn is 1 to about 10. Preferred among these polyethers are iinear and cyclic polyethylene glycols (PEGs), and (PEG)-containing groups, such as crown ethers and, inter alia, those which are disclosed by Delgardo et. ai. (Critical Reviews in Therapeutic Drug Carrier Systems S 992, 9:249), which is hereby incorporated by reference in its entirety. Further sugar modifications are disclosed by Cook {A i-fibrosts Drug Design, 1991 , 6:585-607), Fluoro, Q-alkyl, Q~atkylamino, O-alkyl imidazole, G-aikylarninoalkyl, and aSkyl amino substitution is described in U.S. Patent 6, 166, 397, entitled "Oligomeric Compounds having Pyrimidine NucSeotide(s) with 2' and 5' Substitutions," hereby incorporated by reference in its entirety. Additional sugar substituent groups amenable to the invention include 2 -S and 2 - R2 groups, wherein each R is, independently, hydrogen, a protecting group or substituted or unsubststuted alkyl, alkenyk or alkynyh 2 -S Nucleosides are disclosed in U.S. Pat. No, 5,670,633, hereby incorporated by reference in its entirety. The incorporation of 2 -SR monomer synthons is disclosed by Hamm et al. (J. Org, Chem,} 1 97, 62:3415-3420). 2 -NR nucleosides are disclosed by Goett gers, M, J, Org. Chenh, 1996, 61, 6273-6281 ; and Polushin et al. Tetrahedron Lett., 1 96, 37, 3227-3230, Further representative 2 -substituent groups amenable to the invention include those having one of formula I or II:
Figure imgf000029_0001
II wherein,
E is
Figure imgf000029_0002
{<¾)( ¾); each % and Q4 is. Independently, H5 C Cio alkyl, dialkylansinoalkyl, a nitrogen protecting group, a tethered or untethered conjugate group, a linker to a solid support; or <¾ and <¾, together, form a nitrogen protecting group or a ring structure optionally including at least one additional heteroatom selected from N and O; q3 is an integer from i to 1 ;
¾ Is an integer from 1 to 10;
q4 is Ot l or 2; each Z Z2 arid Z3 is, independently, C4-C7 eycloaikyl,
Figure imgf000029_0003
aryi or C Qs eterocyclyi, wherein the heteroatom in said heterocyclyl group is selected from oxygen, nitrogen and sulfur;
Z4 is OMj, S 3> or N(Mj)25 each M\ is, independently. H, C{-(¼ alkyi, C3-Cs haloalkyl, C(»NH)N(H)M2, C(0)N(H)M2 or OC( ))NiH)M2; M2 is H or C Cg alkyl; and
Zj is Ct-C- alkyl Cj -CJO haioalkyi, C2-C10 alkenyl, C2-CK1 alkynyl, €¾-Cs aryi, (<¼)(<&.), OQ3, halo, SQ3 or CN.
Representative 2'-0-sugsr substituent groups of formula I are disclosed in U.S. Pat No, 6,172,209, eruilled "Capped 2 -Oxyethoxy Oligonucleotides," hereby incorporated by reference in its entirety. Representative cyclic 2 '-O-sugar substituent groups of formula II are disclosed in U.S. Patent 6,271,358, entitled "UNA Targeted 2'-Madified Oligonucleotides that are Conforrnationaliy Preorganized," hereby incorporated by reference in its entirety. Sugars having O-suhstitutions on the ribosyl ring are also amenable to the invention. Representative substitutions for ring O Include, but are not limited to, S, C¾, CHF, and CF2,
Oligonucleotides may also have sugar ndrnetics, such as cydohutyi moieties, In place of the pentofuraraosyl sugar. Representative United States patents relating to the preparation of such modified sugars Include, but are not limited to, U.S. Pat, Nos, 5,359,044; 5,466,?§6; 5,51 , 134; 5,591,722; 5,597,909; 5,646,265 and 5,700,920, ail of which are hereby incorporated by reference.
Additional modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide. For example, one additional modification of the iigand-conjugated oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more additional non~iigand moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide. Such moieties include hut are not limited to lipid moieties, such as a cholesterol moiety (Letsinger et aL, Froc. Natl. Acad. Sci. USA, 1 89, 86, 6553), cholic acid (Manoharan et at., Sioorg. Med Chem, Lett, 1 94, 4, 1053), a thloether, e.g., hexyl~S ritylthiol (Manoharan et ah, Ann, NY. Acad Sc , 1 92, 660, 306; Manoharan et a!., Sioorg. Med Chem. Let, 1993, 3, 2765), a thiocholesterol (Oberhauser et ah, N cL Acids Res., 1992, 20, 533), an aliphatic chain, e.g., dodecandlol or undecy! residues (Saison-Behmoaras et ah, EMBO J., 1991, 10, 1 1 1; Kabanov et ah, FEES Lett, 1990, 259, 327; Svinarehuk et ah, Bioekimie, 19935 75, 49), a phospholipid, e.g., di~hexadecyi~ rac giycerol or triethylarnrnoniurn l,2-di~0-hexadecyi-rac-glycero-3-H-phosphonate (Manoharan et ah, Tetrahedron Lett, 1995, 36, 3653 ; Shea et ah, NucL Acids Res,, 1990, I S, 3777), a poSyarnine or a polyethylene glycol chain (Manoharan et al, Nucleosides & Nucleotides, 1 95, 14, 969), or adamantane acetic acid (Manoharan et ah, Tetrahedron Leu., 1 95, 36, 3651), a palmityl moiety (Mishra et aL, Biochim. Biop ys. Acta, 1 95, 1264, 229). or an octadec lamine or hexylamino-carbon l-oxycholesterol moiety (Crooke et aL, I Pharmacol Exp. Ther., 1996, 277, 923).
The invention also Includes compositions employing oligonucleotides that are substantially chiraiiy pure with regard to particular positions within the oligonucleotides. Examples of substantially chiraiiy pure oligonucleotides include, but are not limited to, those having phosphorothioate linkages that are at least 75% Sp or Rp (Cook et a!., U.S. Pat. No. 5,587,361) and those having substantially chiraiiy pure (Sp or Rp) alkyiphosphonate, phosphoramldate or phosphoiriester linkages (Cook, U.S. Pat Nos. 5.212,295 and 5,521,302). In certai instances, the oligonucleotide may be modified by a non-ligand group. A number of non-Hgand molecules have been conjugated to oligonucleotides in order to enhance the activity, cellular distribution or cellular uptake of the oligonucleotide, and procedures for performing such conjugations are available in the scientific literature. Such non-Hgand moieties have included lipid moieties, such as cholesterol (Letsinger et at., Proc. Nad. Acad. Sci USA, 1989, 86:6553), choiie acid (Manoharan et a!,, Bioorg, Med. Chem. L , 1994, 4: 1053), a thioether, e.g., hexyl-S-trityithioS (Manoharan et al, Aim. NY. Acad Sci., 1992, 660:306; Manoharan et at., Bioorg. Med Chem. Iei.s 1993, 3:2765), a thiocholesteroS (Oberhauser et al, Nucl Acids R s., 1992, 20:533), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et aL, EMEO J,, 1991 , 10: 1 I I ; abanov et al., FEBS Lett.,. 1990, 259:327; Svinarchuk et aL, Biochimie, 1993, 75:49), a phospholipid, e.g., di-hexadecyl-rac-glyeerol or triethylammonium l,2-di-0-hexadeeyl-rac-g!yeero-3-H-phosphonate (Manoharan et al, Tetrahedron Lett, 1995, 36:3651 ; Shea et al., Nnd. Acids Res,, 1990, 18:3777), a polyamine or a polyethylene glycol chair* (Manoharan et al, Nucleosides & Nucleotides, 1995, 14:969), or adamantane acetic acid (Manoharan et al, Tetrahedron Lett^ 1995, 36:3651), a palmltyl moiet (Mishra et al, Biochim. Biophys. Acta, 1995, 1264:229), or an oetadecylamine or hexylarnino- earbonyl-oxycholesteroi moiety (Crooks et al, J. Pharmacol Exp. Ther,, 1996, 277:923), Typical conjugation protocols involve the synthesis of oligonucleotides bearing an aminolinker at one or more positions of the sequence. The amino group is then reacted with the molecule being conjugated using appropriate coupling or activating reagents. The conjugation reaction may be performed either with the oligonucleotide still hound to the solid support or foliowing cleavage of the oligonucleotide in solution phase. Purification of the oligonucleotide conjugate by BPLC typically affords the pure conjugate.
Alternatively, the molecule being conjugated may be converted into a building block, such as a phosphora idite, via an alcohol group present in the molecule or by attachment of a linker bearing an alcohol group that tnay be phosphory!ated.
Importantly, each of these approaches may be used for the synthesis of ligand conjugated oligonucleotides. Amino linked oligonucleotides may be coupled directly with ligand via the use of coupling reagents or foliowing activation of the ligand as an NHS or pentfluorophenolate ester. Ligand phosphorarnidites may be synthesized via tbe attachment of an aminohexanol linker to one of the carboxyl groups followed by phosphitylation of the terminal alcohol functionality. Other linkers, such as cysteamine, may also be utilized for conjugation to a chioroacetyl linker present on a synthesized oligonucleotide.
The person skilled in the art is readily aware of methods to introduce the molecules of this invention into cells, tissues or organisms. Corresponding examples have also been provided in the detailed description of the invention above. For example, the nucleic acid molecules or the vectors of this invention, encoding for at least one strand of the inventive dsKNAs may be introduced into cells or tissues by methods known in the art, like transfeetions etc.
Also for the introduction of dsRNA molecules, means and methods have been provided. For example, targeted delivery by glycosylated and folate-modifsed molecules, including the use of polymeric carriers with ligands, such as galactose and lactose or the attachment of folic acid to various maeromoieeules allows the binding of molecules to be delivered to folate receptors. Targeted delivery by peptides and proteins other than antibodies, for example, including GD- modified nanoparttcies to deliver siRNA in vivo or multieonsponent (nonvlral) delivery systems including short eyelodextrins, adamaniine-PEG are known, Yet, also the targeted delivery using antibodies or antibody fragments, incliiding (monovalent) Fab-fragments of an antibody (or other fragments of such an antibody) or single-chain antibodies are envisaged, injection approaches for target directed deliver)' comprise, inter alia, hydrodynamlc i,v. injection. Also cholesterol conjugates of dsRNA may be used for targeted delivery, whereby the conjugation to lipohilte groups enhances ceil uptake and improve pharmacokinetics and tissue biodlstribution of oligonucleotides. Also cationic delivery systems are known, whereby synthetic vectors with net positive (cationic) charge to facilitate the complex formation with the polyanionic nucleic acid and interaction with the negatively charged cell membrane. Such cationic deliver)' systems comprise also cationic liposomal delivery systems, cationic polymer and peptide delivery systems. Other delivery systems for the cellular uptake of dsRNA/siR A are aptamer-ds siRNA. Also gene therapy approaches can he used to deliver the inventive dsRNA molecules or nucleic acid molecules encoding the same. Such systems comprise the use of non-pathogenic vires, modified viral vectors, as well as deliveries with nanoparticles or liposomes. Other delivery methods for the cellular uptake of dsRNA are extracorporeal, for example ex vivo treatments of cells, organs or tissues, Certain of these technologies are described and summarized in publications, like Akhtat (2007), Journal of Clinical investigation 1 17, 3623*3632, Nguyen ei αί (2008), Current Opinion in Moleculare Therapeutics 10, 138-167. Zamhoni (2005), Clin Cancer Res 1 1, 8230-8234 or Ikeda ei ί. (2006)s Pharmaceutical Research 23, 1631-3640 Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, suitable methods and materials are described below. AH publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict the present specification, including definitions, will control In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
The above provided embodiments and items of the present invention are now illustrated with the following, non-limiting examples.
Description of figures and a ended tables:
Table 1 - Core sequences of dsRNAs targeting human FLT3 gene Letters in capitals represent RNA nucleotides.
Table 2 a) Characterization of dsRNAs targeting human FLT3: Activity testing with single dose. Letters an capitals represent RNA nucleotides, lower case letters "c", "g", "a" and "u" represent 2' Q- eihyi-rnodsiied nucleotides, V represents phosphorothioate and "dT" deoxythyrddine. S.d ~ standard deviation, mRNA ~ mean tnRNA knockdown.
Table 3 -Characterization of dsRNAs targeting human FLT3: Activity testing far dose response. IC 50; 50 % inhibitory concentration, 1C SO: SO % inhibitory concentration, 1C 20: 20 % inhibitor)' concentration.
Table 4 - Characterization of dsRNAs targeting human FLT3: Stability and Cytokine Induction, t ½ : half-life of a strand as defined in examples, PBMC; Human peripheral blood mononuclear cells. able 5 - Core sequences of dsRNAs targeting human FLT3 gene and their modified counterparts. Letters in capitals represent RNA nucleotides, lower ease letters "c55, "g", "a" and "u" represent 2' O-methyl-niodif ed nucleotides, "s" represents phosphorothioate and "dT" deoxythyrnldine. Table 6 - Sequences of bDNA probes for determination of human FLT3. LE- label extender, CE- capture extender, BL= blocking probe.
Table 7 - Sequences of bDNA probes for determinatio of human GAPDH, LE~ label extender, CE= capture extender, BL~ blocking probe.
Table 8 - Selected off-targets of dsRNA targeting FLT3, SEQ ID NO pair 295/296,
Table 9 - Selected off-targets of dsRNA targeting FLT3, SEQ ID NO pairs 167/168 and 297/298
Table 10 - Selected off-targets of dsRNA targeting FLT3? SEQ ID NO pair 293/294. Table 11 - Sequences of bDNA probes for determination of reduction in off-target expression by dsRNA SEQ ID NO pair 295/296. LE™ label extender, CE~ capture extender, BL™ blocking probe.
Table 12 - Sequences of bDNA probes for determination of reduction i off-target expression by dsR A SEQ ID NO pairs 167/168 and 297/298. LE= label extender, CE- capture extender, BE51 blocking probe
Table 13 - Sequences of bDNA probes for determination of reduction in oil-target expression by dsR?<IA SEQ ID NO pair 293/294. LE= label extender. CE= capture extender, BL~ blocking probe
Table 14 ~ Sequences of bDNA probes for determination of human GAPDH by in vitro off-target analysis. L£;:: label extender, CE capture extender, BL= blocking probe.
Figure 1 - Off-target analysis of selected off-targets of dsRNA SEQ SD NO pair 295/296: Single Dose Analysis in Hela Cells [50 nM]
Fi ure 2 ~ Off-target analysis of selected off-targets of dsRNA SEQ ID NO pair 295/296: Dose response curves Fi u e 3 - Off-target analysis of selected off-targets of ds NAs SEQ ID NO pairs
167/168 and 297/298: Single Dose Analysis in Hela Cells [50 n j Fi ure 4 - Off-target analysis of selected off-targets of dsRNA SEQ I'D NO pair 167/168 and 297/298: Dose response curves
Figssre S - Off-target analysis of selected off-targets of dsRNA SEQ ID NO pair 293/294: Single Dose Analysis in Mela Cells [50 rsM]
Figure€ ~ Dose-dependent FLT3 mRNA knockdown in Moiml3 ceils after 24 hr transfection,
Flgssre 7 - Dose-dependent FLT3 mRNA knockdown in oIml3 cells after 24 hr transfeetion: Decrease of the immediate downstream STATS phosphorylation. Control siRNA; ETO (SEQ ID, NO pair 348/349)
Flgis re S - Dose-dependent growth inhibition by FLT3 dsRNAs (5 days).
Fi ure 9 - Cell cycle analysis after 24 hr transfeetion of Molm 13
Figure imgf000035_0001
d$RN A design was carried out to identify dsRNAs specifically targeting human FLT3 for therapeutic use. First, the known mRNA sequence of human (Homo sapiens) FLT3 (NM_O041 19,2 listed as SEQ ID NO. 1 50) was downloaded from NCBI Genbank.
The eynomolgous monkey (Macaea fascicularis) FLT3 gene was sequenced (see SEQ ID NO. 1622) The cyrsomolgus monkey sequence (SEQ ID NO. 1651) was examined together with the human FLT3 mRNA sequence (SEQ ID NO, 1650) by computer analysis to identify homologous sequences of 19 nucleotides that yield RNA interference (RNAi) agents eross-reaetive to both sequences.
In identifying RNAi agents, the selection was limited to 19mer sequences having at least 2 mismatches lo any other sequence in the human RefSeq database (release 3S}} which we assumed to represent the comprehensive human iranseriptome, by using a proprietary algorithm. All sequences containing 4 or more consecutive G's (poly-G sequences) were excluded from the synthesis,
The sequences thus identified formed the basis for the synthesis of the RNAi agents in appended Tables 1, 2 and 5. dsRNAs cross-reactive to human as well as cynomoigoiis monkey were defined as most preferable for therapeutic use.
dsUNA s hm
Where the source of a reagent is not specifically gi en herein, such reagent may be obtained from any supplier of reagents for molecular biology at a quality/purity standard for application Irs molecular biology.
Oligoribonucleotides were assembled on an AB1 3900 synthesizer (Applied Biosystems) according to the phosphoramsdite oiigornerlzation chemistry. The solid support was polystyrene loaded with 2'~deoxy~thyrnldine (purchased from Glen Research, Sterlings Virginia, USA) to give a synthesis scale of 0.2 μηιοΐ. Ancillary synthesis reagents, RNA and 2'- -Methy! NA phosphoramidiies were obtained from SA C Proilgo (Hamburg, Germany). Specifically, S'-O- (4,4'-dimethoxytrityl)~3 '-0-(2-cyanoethyi-jV,iV-diisopi'opyl) phosphorarnidite monomers of uridine (U)f 4«N-acetyleyiIdine (C 6-N~benzoyIadenosine (Ate') and 2-N sobutyr!guanosine (GiBlJ) with 2'-0-/-biityIdimethylsiiyl were used to build the oligomers sequence. 2'-0- ethyl modifications were introduced employing the corresponding phosphoramidiies carrying the same nucieobase protecting groups as the regular RNA building blocks. In order to generate a 5*~ phosphate the 2-[2-(4,4,~Dime hox} rityl xy)ethylsulfonyllethyi-(2»cyanoethyl) VsN- diisopropyi)-phosphoramidite from Glen Research (Sterling, Virginia, USA) was used. Coupling time for ail phosphoramidites (70 mM in Acetonitriie) was 3 min employing 5-EthylthIo-I H- tetrazole (ETT) as activator (0.5 M In Acetonitriie), Phosphorothloate linkages were Introduced using 50 mM 3 (DImethySaniIno~methylideiie)amIno)-3H-l s2f4-dithIazoie~3-thione (DDTT, AM Chemicals, Oceanside, California, USA) in a 1 ; 1 (v/v) mixture of pyridine and Acetonitriie, Upon completion of the solid phase synthesis oHgorlbonucleotides were cleaved from the solid support and deprotected using slight modification of published methods (WincottF. et si. Synthesis,, deproteeiion, analysis and purification of RNA and ribozymes. Nucleic Acids Res., 1995, 23, 2677-2684), Subsequently, the crude oligoribonucieotldes were purified by anion- exchange high-performance liquid chromatography (HPLC) on a AKTA Explorer System (GF. Healthcare, Freiburg, Germany), Purified oiigorihonucleotldes were desalted by size exclusion chromatography employing a HiTrap 5 niL column (GE Healthcare). Identity of the oligoribomseleotides was confirmed by MALDi mass spectrometry and purity was assessed by 5 analytical anion-exchange HPLC, To generate siRNAs from RNA single strands, equimolar amounts of complementary sense and antisense strands were mixed and annealed in a 20 mM NaCi, 4 mM sodium phosphate pH 6,8 btrf er, siRNAs were further characterized by capillary gel electrophoresis and were stored f oz until use.
ί 0 The activity of the FLT3~dsRNAs for therapeutic use described above were tested in four different assay systems. One approach uses MOLM13 and MV4-1 1 ceils, electroporated with siRNAs, In a second approach a DuaiGlo reporter-assay system was used. For this the human FLT3 sequence was cloned into psiCHEC ™-2 Vector (Fromega GmbH Mannheim, Germany, cat-No CS02I) expressing Firefly- and Renilla-Luciferase. MCF7 and TC7I cells, expressing i 5 FLT3 endogenous!)', were used in a third approach. In a further approach MOLM13 cells were treated with siRNAs formulated in LNP (internally prepared Lipid Nano Particles).
Cell Culture: MOLM13 ceils were obtained from DSAfZ Deutsche Sammlung von Mikroorganlsmen und Zelikulturen GmbH (Braunschweig, Germany, cat No, ACC-554) and 0 cultured in suspension in RP I 1640 (Bioehrorn AG, Berlin, Germany, cat. No FG 1215)
supplemented to contain 10% fetal calf serum (FCS) (Biochrom AG, Berlin, Germany, eat. No. S01 15), 1% Penicillin / Streptomycin (Gibco Invitrogen, cat. No.15140-122) at 37°C in an atmosphere with 5% C02 in a humidified incubator (H eraeus HEf¾.AcslL K ndro Laboratory Products, Langenselbold, Germany). COS7 cells were obtained from DSMZ Deutsche Sammlung 5 von Mikroorganismen und Zelikulturen GmbH (Braunschweig, Germany, cat No. ACC-60) and cultured m DMEM (Biochrom AG, Berlin, Germany, cat. No. F0435) supplemented to contain 10% fetal calf serum (FCS) (Gibco Inviirogen catNo.16250-078), 1% Penicillin / Streptomycin (Gibco Invitrogen, cat No.15140- 122), 5 ml L-Glutamin (Biochrom AG, Berlin, Germany, cat No 0283) and 5 mi 1 ,2 mg mi Natriumbicarbonat (Biochrom AG, Berlin, Germany, cat No L0 1703) at 3 C in an atmosphere with 5% C02 in a humidified incubator (Heraeus HERAceil, endro Laboratory Products, Langenselbold, Germany), MCF7 cells were obtained from DSMZ Deutsche Sammlung von Mikroorganismen und Zelikulturen GmbH (Braunschweig, Germany, cat No. ACC 1 15) and cultured Irs RPMI 1640 (Biochrom AG, Berlin, Germany, cat. No. FG 1215) su lemented to contain 10% fetal calf serum (FCS) (Gibco Invitrogen catNo.16250-078), 1% Penicillin / Streptomycin (Gibco nvitrogen, cat No.15140-122), 5 ni3/50GmI EAA (nonessential aminoacids) (Biochrom AG, Berlin, Germany, cat No 0293), 5 ml/SGOml
Natriumpyr vat (Biochrom AG, Berlin, Germany, cat NoL 0473), and !Qpg/ml human insuHne (Sigrna-Aldrieh, Germany, cat No I0516-5ML ) at 37°C is an atmosphere with 5% C02 In a humidified incubator (Heraeus HERAcell, Kendro Laboratory Products, Langenselboid,
Germany). TC71 cells were obtained from DSMZ Deutsche Sammlimg von Mikroorganismen und Zellkiiituren GmbH (Braunschweig, Germany, cat. No, ACC516) and cultured in I DM (Gibco Invitrogen, Germany, cat. No. 21980-032) supplemented to contain 10% fetal calf serum (FCS) (Gibco inviirogen cat.No.16250-078), and 5ml/500mS Penicillin / Streptomycin (Gibco Inviirogen, cat. No.15140-122), at 37 in an atmosphere with 5% CO! in a humidified incubator (Heraeus HERAcell, Kendro Laboratory Products, Langenselboid, Germany). MV4-1 1 ceils were obtained from DSMZ Deutsche Sammlung von Mikroorganismen und ZeHkulturen Grnbl l (Braunschweig, Germany, cat No. ACC 102) and cultured sn RPMI 1640 (Biochrom AG? Berlin, Germany, cat, No. FG 1215) supplemented to contain 10% fetal calf serum (FCS) (Gibco Invitrogen catNo.16250-078), and 5ml/50Qml Penicillin / Streptomycin (Gibco Invitrogen, cat. No.15140-122), at 37aC in an atmosphere with 5% C02 in a humidified incubator (Heraeus HERAcell Kendro Laboratory Products, Langenselboid, Germany).
L Electroporation of MOLM13 cells and MV4-11 ceils: Efficient introduction of FLT3 targeting siRNA into suspension cells were performed with either Neon Transfection System (Gibco Invitrogen, cat. No, MP 5000) or GneDrop Mieroporator MP- 100 System (BTX/ Harvard Apparatus) following the manufacturer's protocol Three different concentrations of siRNA (!OOnM, 250nM and SOOnM) were used for single dose experiments. In a volume of 10 μΙ 3 times 10 sup,4 ceils were dectroporated with the appropriate amount of siRNA and plated in 96-weIl plate containing ΙΘΟμΙ prewarmed cell-medium. 4 h alter electroporation ceils werde collected for NA quantification. bDNA Assay Kit QuantiGene 2,0 (Pano ics/ ffymetrix, Fremont, USA, Cat-No: 15735) was used for quantification of FLT3 mRNA, while QuantiGsne Assay
1.0 iPanomics/Affytnetrix, Fremont, USA, Cat-No: QG0004) was used for quantification of GAPDH mRNA, Cells were harvested and lysed at 53CC following procedures recommended by the manufacturer Panomics/Affymetrix for bDNA quantitation of rrjRNA. Afterwards, 50 μΐ of the iysa es were incubated with probeseis specific to human FLU and human GAPDH (sequence of probeseis see below) and processed according to ihe manufacturer's protocol for QuantlGene Assay Kit 1 or 2, respectively,
ChemQiumineseence was measured in a YIctor2-Light (Perkin Elmer, Wiesbaden, Germany) as RLUs (relative light units) and values obtained with the human KIF10 probeset were normalized to the respective human GAPDH values for each well Unrelated control siRNAs were used as negative control,
DualG!o-Reporter assay: This assay was performed in a 96-weil plate formal.
Lipofectamine 2000 (Invitrogen GmbH, Karlsruhe, Germany, cat.No. 11668-01 was used as transfection reagent following the protocol of the manufacturer. Ceil seeding and iransfection of plasmid-DNA (SOng well) were performed at the same time. Transfection of siRNA was carried out 4 h later. For transfection COS7 cells were seeded at a density of 2.5 times 10„sup,4 cells/well. In a first single dose experiment siRNAs were trausfeeted at a concentration of 50 nM. Most effective siRNAs against FLT3 from the single dose screens at 50n were further characterized by dose response curves. For dose response curves, transactions were performed as described above for the single dose screen, but with the following concentrations of siRNA (nM): 24, 6, 1 ,5, 0,375, 0.0938, 0,0234, 0.0059» 0.0015, 0.0004 and 0.0001 nM . After transfection cells were incubated at 3?°C and 5 % C02 is a humidified incubator (Heraeus HE Acell, endro Laboratory Products, Langenselbold, Germany). 24 hours after transfection ceils were analyzed for the expression of Renilia and Firefly-Luciferase using the DualGlo Kit (Promega) according to the protocol described by the manufacturer. Luminescence was measured in a Vietor2~Light (Perkin Elmer, Wiesbaden, Germany) as RLUs (relative light units) values obtained for RenIHa Luciferase were normalised to the Firefly Luciferase values for each well Unrelated control siRNAs were used as a negative control.
MCF7 cells: Cell seeding and transfection of siRNA were performed at the same time. For transfection with siRNA, cells were seeded at a density of 2.5 times 10 sup.4 cells/well in 96-well plates. Transfection of siRNA was carried out with lipofectamine 2000 (.Invitrogen GmbH, Karlsruhe, Germany, ca .No. 1 1668-019) as described by the manufacturer. In a first single dose experiment siRNAs were transacted at a
concentration of 50 nM. in a second single dose experiment most active siRNAs were reanalyzed at 30pM Most effective siRNAs against IF10 from the single dose screen at 30 pM were further characterized by dose response curves. For dose response curves, transtections were performed as described for the single dose screen above, but with the following concentrations of siRNA (nM): 24, 6, 1.5, 0.375, 0.0938, 0.0234, 0.0059, 0.0015, 0.0004 and 0.0001 nM . After transfeetion ceils were incubated at 37*C and 5 %
C02 in a humidified incubator (Herae s HERAceli, endro Laboratory Products, Langenselbold, Germany), 24 h after electroporation ceils werde collected for RNA quantification using bD A Assay Kit as described above, 4. Transaction of OLM13 cells with L P formulated siRNAs: Cells were plated in 6- weil plates at I x S O sup.6 cells/well and mixed with LNP containing the indicated amounts of FLT3 targeting siRNA at concentrations of l OGnM and 40nM. After 1 8 hours of transfeetion, the medium was changed to the respective cuhurmg medium. Cells were collected for RNA quantification using RT-PCR, Total RNA was purified using Qiagen RNeas Kit (Qiagen, Hilden, Germany, cat, No 7 106) following the manufacturer's protocol Relative quantification of FLT3 and 18S ribosomal mRNA was carried out with High Capacity cDNA Reverse Transcription Kit {Applied Biosystems) followed by Taqrnan Gene Expression Assays using the manufacturer's protocol, (Applied
Biosystems, eat No 4319413E),
Inhibition data are given in appended tables 2 and 3. Stability of dsRNAs
Stability of dsRNAs targeting human FLT3 was determined in in vitro assays with either human or mouse serum by measuring the half-life of each single strand. Measurements were carried out in triplicates for each time point, using 3μΙ 50μΜ dsR A sample mixed with 30μ1 human serum (Sigma) or mouse serum (Sigma). Mixtures were incubated for either Gmin, 3 Grain, Ih, 3h, 6h, 24h„ or 4Sh at 37*C. As control for unspecific degradation dsR A was incubated with 30μ1 Ix PBS pH 6.8 for 48h. Reactions were stopped by the addition of 4μ1 proteinase K (20rng/ml), 25μ! of ' issue and Cell Lysis Solution" (Epicentre) and 38μ1 Miliipore water for 30 min at 65°C. Samples were afterwards spin filtered through a 0.2 μπι 96 well filter plate at 1 00 rpm for 8 min, washed with 55μ! Miilipore water twice and spin filtered again. For separation of single strands and analysis of remaining full length product {FLP), samples were run through an Ion exchange Dionex Summit I IPLC under denaturing conditions using as eluent A 20mM Na3P04 in 10% AC'N pH=l 1 and for eiuent B 1 M aBr in eluent A.
The following gradient was applied;
Figure imgf000041_0001
For every injection, the chromaiograms were integrated automatically by the Dionex Cliromeieon 6.60 HPLC software, and were adjusted manually if necessary. Ail peak areas were corrected to the internal standard (IS) peak and normalized ΐο the incubation at t=0 min. The area under the peak and resulting remaining FLP was calculated for each single strand and triplicate separately. Half-life (tl/2) of a strand was defined by the average time point [h] for triplicates at which half of the FLP was degraded. Results are given in appended table 4,
Potential cytokine induction of dsRNAs was determined by measuring the release of !NF-a and TNF-a in an in vitro PBMC assay. Human peripheral blood mononuclear ceils (PBMC) were isolated from buffy coat blood of two donors by Ficoil eentrifugation at the day of transfection. Cells were transfected in quadruplicates with dsRJNA and cultured for 24h at 37CC at a final concentration of DQrsM in Opts-MEM, using either Gene Porter 2 (GP2) or DDTAP. dsRNA sequences that were known to induce F F~a and TNF-a in this assay, as well as a CpG oligo, were used as positive controls. Chemical conjugated dsRNA or CpG oligonucleotides that did not need a transfection reagent for cytokine induction, were incubated at a concentration of 500nM in culture medium. At the end of incubation, the quadruplicate culture supernatant were pooled,
I F-a and T P-a was then measured in these pooled supernatant? by standard sandwich ELISA with two data points per pool. The degree of cytokine induction was expressed relative to positive controls using a score from 0 to Ss with 5 indicating maximum induction, Results are given in appended table 4.
Cell eisitisre md siRNA ir&Bsffeetiesss, HepG2, HLF and A549 ceils were obtained from ATCC and maintained in the recommended media, supplemented with 10% fetal bovine scrum and 2mM 1-Glutamine (HepG2: MEME; HLF and A549: DMEM). Ceils were trarssfected using 0J μΐ DharmaFeci 3 (Thermo Fisher) per well of a 96-weSl plate, with each well containing a final volume of 100 μί growth media. Transfections in 6-weIl plates were carried out in a similar manner, with volumes adjusted for the larger well size. Transfections were performed using a "reverse transfection" protocol, in which cells (HepG2: 5,000 cells; HLF: 2,000 celis; A540: 4,000 cells) were mixed with transfection mix immediately prior to plating,
Figure imgf000042_0001
Iss s lko off-tergei prediction
The human genome was searched by computer analysis for sequences homologous to the inventive dsRNAs. Homologous sequences that displayed less than 6 mismatches with the inventive dsRNAs were defined as a possible off-targets. Off-targets selected for in vitro off target analysis are given in appended tables g-iO.
Analysis of dsK A off-target effects
Analysis of silencing putative off-targets was performed after transfection of either human HeLa cells or human A431 cells using Lipofectamine 2000 (Invitrogen GmbH, Karlsruhe, Germany, cat. No, 1 1668-019). A431 and cells were obtained from Deutsche Santmlung fur Mikroorganismen und ZellkuSturen (DSMZ, Braunschweig. Germany, cat. No. AOC-9I) and cultured in RPMI (Biochrom AG, Berlin, Germany, cat. No. FG 1215) supplemented to contain 10% fetal calf serum (FCS) (Biochrom AG, Berlin, Germany, cat. No. S01 15), Penicillin 100 U/ml, and Streptomycin 100 pg/ml (Biochrom AG, Berlin, Germany, cat. No. A2213) at 37°C in an atmosphere with 5% C02 in a humidified incubator (Heraeus HERAceii, endro Laborator Products, Langenselbold, Germany). Heia cells were also obtained from Deutsche Sammiung fur Mikroorgantsmen und Zellkulturen (DSMZ, Braunschweig, Germany, cat. No. ACC-57) but cultured under different conditions (MEM with Earie's salt - ATCC 30-2003 supplemented with 10% fetal calf serum [FCS] [Biochrom AG, Berlin, Germany, eat. No. SO 115], Penicillin 100 U/ml, and Streptomycin 100 u-g rnl [Biochrom AG, Berlin, Germany, cat. No. A2213] as well as 2 mM L-Glutamine [Biochrom AG, Berlin, Germany, cat, No, 02S3] at 3?°C hi an atmosphere with 5% C02 is a humidified incubator. After transfeetion cells were incubated for 24 h at 37 °C and 5 % C02 in a humidified incubator (Heraeus GmbH, Hanau, Germany), For measurement of all off target niRNAs as well as GAFDH m NA the QuantiGene 2,0 Assay Kit (Panomies, Fremont, Calif,, USA) for bDNA quantitation of mRNA was used. Transfected A431 and/or Hela cells were harvested and lysed at 53 °C following procedures recommended by the manufacturer. 20 μΐ of the lysates were incubated with prohesets specific for human off target mRNAs (sequence of probesets see Table 1 1 to 13, SEQ ID Mo 350-1629 } and processed according to the manufacturer's protocol for QuantiGene, For measurement of GAFDH mRNA 3 μϊ of the ceil lysate was analyzed with the GAPDH specific probeset (Fable 14, SEQ ID No 1630-1649). Chemoinmineseenee was measured in a Vicior2~Light (Ferkln Elmer, Wiesbaden, Germany) as RLUs (relative light units) and valises obtained with the human off target probeset were normalized to the respective human GAPDH values for each well. Unrelated control siRNAs were used as a negative control. All data regarding the off-target analyses are given in tables 8-10 and figures 1 -5.
Figure imgf000043_0001
The human cancer cell line MOLM 13 (DSMZ, Braunschweig, Germany) was maintained in media supplemented with 10% heat-inactivated Fetal Bovine Serum (HI-FBS; GlBCO BRlL Gaithersburg, MD) and 2 mM L-glutamine (GIBCO/BRL),
Tnmsfeeilon
2 U)5 MoirnD cells in 1 nil of culiuring medium were seeded in 24-well plates, FLT3 siRNAs formulated with internal LNP were mixed as indicated concentration for 24 hours , Ceils were collected ibr RNA quantification.
RT-PCR
Sample collection and mRNA purification for in vitro studies were performed as follows. Suspension cells were collected into tubes and spun down at 2,000 rpm for 1 min, then the cell pellets were lysed w th RNA lysis buffer (Qiagen), Total RNA from all collected samples was purified using Qiagen RNeasy Kit following the manufacturer s protocol Relative quantification of FLT3 mRNA and 18S ribosoraal RNA gene expression was carried out with cDNA Reverse Transcription Reagents from Applied Biosystems followed by Taqman Gene Expression Assays (Applied Biosystems) using the manufacturer's protocol. The catalog numbers for each probe set were: human FLT3 (Hs00975659_ml) and 18S (431941 E).
Westers blot analysis
1.2XI06 Mohn l 3 ceils in 3 ml of culturing medium were seeded in 6-weil plates, FLT3 siRNAs formulated with internal LNP were mixed as indicated concentration for 24 hours . Cells were collected into tubes and spun down at 2,000 rpm for 1 min, then the ceil pellets were lysed with protein lysis (2,5% SDS, 10 raM Tris pH7.5, 2 mM EDTA) Proteins were denatured by boiling for 5 minutes. Protein concentration were measured by BCA protein assay kit (Therrno Fisher), Equal amount of protein samples was resolved by SDS-polyacr lamide gel electrophoresis using a 4-20% Tris-giyeine gel (Invitrogen) and electroblotted onto a 0.45 μι¾ nitrocellulose membrane (invitrogen). Membranes were blocked S hr at room temperature in blocking buffer (5% milk in PBS/0.1% Tween 20) followed by incubation with the primary antibody at 4°C overnight. Membranes were washed and incubated with the secondary antibody for 30 minutes at room temperature, Immunodetection was carried out using enhanced chemolnminescence (ECL Plus, Araersbam Pharmacia Biotech, Piseataway, NJ). For Western blotting, FLT3 was detected using the FLT3 aniibody from Cell Signaling (#3462) at a dilution of 1 :500, STATS phosphorylation was detected using the antibody from Cell Signaling (#9358) at a dilution of 1 :500» phospho-STATS was detected using the antibody from Ceil Signaling (#9359) at a dilution of 1 :500, and actln was detected using the actin antibody from Sigma (#5316) at a dilution of 1 : 10,000.
Cell Cycle analysis
Cells were incubated with FLT siRNAs for 24 hours, washed twice in phosphate-buffered saline (PBS), and fixed at -20°C overnight with 70% eihanol. Cells were then analyzed using propidium iodide (Pi) staining (Beoton Dickinson, San Jose, CA), Briefly, cells were washed twice with cold PBS and incubated with Pl/RNase solution (Becton Dickinson, San Jose, CA) for 15 min at 37 . Samples were n l ze on a Acuuri C6 flow cyfometer and its software (Accurl, Ann Arbor, MI). RESULTS
Results are shown in Figures 6-9. Transfeetion of the Mofm-I 3 AML tumor derived ceil line with siRNAs directed towards FLT3 mRNA produced potent mRNA knockdown which correlated with protein knockdown. The phenofype associated with the loss of FLT3 expression is characterized by the loss of STAT5-F, a Gi arrest producing a swb-Gl population of cells and loss of cell viability. These results are consistent with the cellular inhibition of FLT3 signaling.
Table 1
SEQ II
serssc ί I seqssenee (5 '-3'} an&s sse strasd sequenc (S"-3*
UCAGCUAUUUAGUCAUAUA UAUAUCACUAAAUAGCUGA
GGACCAAUUUACUUGAUUU AAAUCAAGUAAAUUGGUCC
GUUUACAGLfGAGUAUAAGA UCUUAUACUCACUGUAAAC
AGUUUUGCAAUCAUAAGCA UGCUUAUGAUUGCAAAACU
AGAGUG A AGCUACC A AG U A UAAUUGGUAGCUUCACUCU
CAUGUUGUGAGACGAUCCU AGGAUCGUCUCACAAGAUG
UGAUGAACGCAACAGCUUA 14 UAAGCUGUUGCGUUCAUCA
I S AUUUAAGUCGUGUGUUCAC GUGAACACACGACLfUAAAU
17 UGUUUUAAUCAAUCAUAAG CUUAUGAUUGAUUAAAACA
GGAUUUAUAAAUGCUACCA UGGUAGCAUUUAUAAAUCC
21 CUGCAUAUCUGAGAGCGUU AACGCUCUCAGAUAUGCAG
23 GGUAUGAAAGCCAGCUACA 24 UGUAGCUGGCUUUCAUACC
25 UACIJUUGAGAUGAGUACCU 26 AGGUACUCAUCUCAAAGUA
27 GUACUUCUACGUUGAUUUC GAAAUCAACGUAGAAGUAC
29 GUAACAUGGAGGAUUAGUA 30 UACUAAUCCUCCAUGUUAC
31 AGGACCAAU UUACUUGAUU 32 AAUCAAGUAAAUUGGUCCU
AACGCAACAGCUUAUGGAA 34 UUCC AUAAGCUG UUGCGUU
35 AGGUUUAAAGCCUACCCAC 36 GUGGGUAGGCUUUAAACCU
CUUGGCACAUCUUGUGAGA UCUCACAAGAUGUGCCAAG
39 AUUGUGUACCUGAAGiJACA 40 UGIJACUUCAGGUACACAAU
41 AGUACUUCUACGUUGAUUU 42 AAAUCAACGUAGAAGUACU
43 ACCAAAACAGGGGACCUUU AAAGGUCGCCUGUUUUGGU
AUCAAGUGUGUUUUAAUCA 46 UGAUUAAAACACACUUGAU
CCAAAACAGGCGACCUUUC GAAAGGUCGCCUGUUIJUGG
GGCACAUCUUGUGAGACGA 50 UCGUCUCACAAGAUGUGCC
GCUCCUCAGAUAAUGAGUA 52 UACUCAUUAUCUGAGGAGC
53 AGUGCUGUGCAUACAAUUC 54 GAAUUGUAUGCACAGCACU
55 GCUUUGGUUACCAUCGUAG CUACGAUGGUAACCAAAGC
57 UACCAAAACAGGCGACCiJU 58 AAGGUCGCCUGUUUUGGUA
59 UAACAGGCUGUAGAUUACC 60 GGUAAUCUACAGCCUGUUA
UUGGUUACCA!JCGUAGAAA UUUCUACGAUGGUAACCAA
GCACAUCUUGUGAGACGAU AUCGUCUCACAAGAUGUGC
UACAUUAUAAUGCAAUCCU 60 AGGAUUGCAUUAUAAUGUA
67 AGGCUGUAGAUUACCAAAA UUUUGGUAAUCUACAGCCU
GCUGGCUUGAGUGAAUUGU ACAAUUCACUCAAGCCAGC
GCUACAGAUGGUACAGGUG 72 CACCUGUACCAUCUGUAGC
73 CUACCAAAACAGGCGAGCU 74 AGGUCGCCUGUUUUGGUAG
75 AACAGAACUAUGAUACGGA 7$ UCCGUAUCAUAGUUCUGUU
77 ACACACGACUUAAAUUCCA
Figure imgf000047_0001
Table 2 a)
Figure imgf000048_0001

Figure imgf000049_0001

Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Activity testin with Activity testing with Activity testing with S M siMNA in MCF? SOiiM s!R A in MCF? SOaM siRNA £» TC7S ee!Ss cells cells
standard mean stan ar mean Standard rcmz'm g deviation remainin deviation remaining deviation
SEQ ID NO pair JHRNA (%i !%i RNA f%| f%I l%1
209/210 46 7,499 48 6.881 46 7.499
21 1/212 55 7.756 57 1 1.341 55 7.756
213/214 59 3.300 67 8.543 59 3.300
215/216 64 10.022 62 13.272 64 10.022
217/218 57 9.339 64 17.603 57 9.339
219/220 65 1 5.575 76 9.059 65 15,575
38 2.997 40 4.207 38 2.997
223/224 74 Ώ.661 87 12.496 74 13,661
225/226 51 1 1.41 1 63 13,018 51 1 1.41 1
227/228 62 1 1.388 76 1 1.132 62 1 1.388
229/230 59 6.676 73 9.386 59 6.676
231/232 62 10.816 74 12,81 1 62 10,816
2.33/234 45 6,374 47 1 1.995 45 6.374
235/236 54 1 1.886 69 8.812 54 1 1 .886
237/238 62 19.145 82 8.612 62 19.145
239/240 36 14.01 1 49 8.441 36 14.011
241/242 68 13.924 74 16.950 68 13.924
243/244 82 13.825 97 12.289 82 13.825
245/246 42 18.747 68 1.734 42 18,747
247/248 57 16.436 68 1 1.188 57 16,436
249/250 75 20.321 101 8.889 75 20.321
251/252 73 12.132 86 1 1.371 73 12.132
253/254 69 5.962 84 15.066 69 5.962
255/256 73 10.584 87 32.273 73 10.584
257/258 53 12.662 65 13.193 53 12.662
259/260 85 12.454 93 24.577 85 12.454
261/262 53 10.591 65 12.378 10.591
263/264 50 13.841 64 10.1 15 50 13.841
265/266 67 23.505 87 9.064 67 23.505
267/268 66 26.720 83 9.744 66 26.720
269/270 74 7.568 77 8.066 74 7.568
271/272 66 17.728 81 13.574 66 17.728 S3
Figure imgf000054_0001
Table 2 c)
Activity iest ig with Activity es in wiils SiHisiM ss NA 883 lOSsiM s A in MOLM cells MOLSV113 eel!s {ekctrofsorsiioss} (eSeciroporation) ean stadard s*seas¾ standard reniasmsig deviation deviation
SEQ m NO pair BiRNA [%S JHRNA I%1 |%|
147/148 42 3.427 60 5,438
149/150 27 4.066 39 0.375
151/152 39 6.753 56 4.553
153/154 38 1.092 65 0.000
155/156 30 1.397 48 2.420
159/160 29 0.798 68 0.977
165/166 52 3.090 91 8.873
167/168 30 4374 57 2.817
171/172 28 0.634 38 10.137
173/174 30 0.533 48 9,441
179/180 46 1.094 84 0,949
1 81/182 33 1 .021 51 3.325
1 83/184 76 8.521 84 3.978
197/198 74 8.650 123 19.421
199/200 92 3.258 104 1.561 Activit tesisag with Activity it stis¾g with
SOOfiM sf A iffi iRNA in MOLM eelis M LM JSeeiis (eleciro oraiion;} {e!eciroj
standard ssseai* siassdar deviation EQ ID NO pir mRNA [%] |%| HKRNA j%|
221/222 81 0.314 115 5.117
277/278 89 11.132 104 7.704
Table 2 d)
Figure imgf000055_0001
55
Figure imgf000056_0001
J Aciiyiiy t« isiiiig with
500s 5 iRNA in
OLJV El 3 eeils
{efeeiroi sorption)
Standard
SEQ m iresnasssing deviation
NO pair MSR A j%| l l
255/256 108 4.916
257/258 82 21.532
259/260 115 7.135
261/262 138 17.253
263/264 88 8.029
265/266 115 15.234
267/268 121 8.087
269/270 128 5.967
271/272 113 16.217
273/274 117 12.214
275/276 100 1.917
277/278 110 1.003
279/280 111 0.000
281/282 86 9,943
283/284 145 9.934
285/286 112 12.693
287/288 124 7.408
289/290 173 8.651
291/292 108 1.579
Table 2 e)
Activity iestifig with Activity iestf!g with SOOK siRNA m !O ss!VI siRNA m MOL 13 ceB!s MOLM13 «eiis (eSectr porsiiosii) (electroporatioii) standard aieaii standa d
SEQ ID de iaiioKS r maining deviation NO pair mRNA |%| l%! iiiR A |%| I%1
147/148 83 6.040 98 0.046
149/150 60 3.090 110 4,990
151/152 50 5.588 153 33.975
153/154 41 10.787 86 10.534
155/156 66 3.573 142 25.999
157/158 38 0.000 49 0.000 Activity testin mth Activity tesiiag with SOOHM SJENA sss 10©HM SSRN.4 in MOLMO cells MOLM13 ceils Cetectro orataogi)
meats standard mean s anda d
SEQ m remaining devsatsoss
NO pair mRNA [%] [%] ΚΪΜ Α {%] ! [%]
1 59/160 57 15.81 1 91 29.469
161/162 45 5.560 78 6.162
163/164 58 1 1.764 156 4.35 !
167/168 36 5.085 63 1 1.679
171/172 1.065 59 7.565
173/174 55 3.049 90 47.008
181 /182 106 7.848 140 15.771
185/186 67 10.953 97 16.894
195/196 76 16.465 120 1.066
203/204 71 0.996 95 3.689
219/220 70 0.483 122 14.971
Figure imgf000058_0001
Figure imgf000059_0001
Table 2 g)
Figure imgf000059_0002
Table 2 h
Figure imgf000060_0001
Table 2 i)
Activity testing with Activity testing with Activity testing with SOOiiM siR A in 25ί>ίίΜ siRNA ίΟ κΜ siR A in MOL 13 eeiis MOLM13 cells MOLM 13 ceils dectro ration) (deetroporation) ] (eiecira oration) meass standard Kicas standard i!ieas standard
SEQ ID NO deviation remaining deviation retrsaimng deviation pair mRNA |%j %| w-R A [%| [ ] mRNA {%] f%]
149/150 28 7 44 5 47 7
151/152 37 5 51 13 67 23
153/154 31 4 47 5 69 8
157/158 23 3 40 5 47 6
159/160 44 8 48 10 63 8
161/162 29 5 39 s 35 6
163/164 79 7 98 8 136 7
167/168 2 5 23 2 24 1
171/172 31 2 37 2 61 l i
173/174 25 10 35 4 50 5 Table 2 )
Figure imgf000061_0002
Figure imgf000061_0001
61
Figure imgf000062_0001
Table 3
Figure imgf000063_0001
'fable 4
Figure imgf000064_0001
64
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Table 6
FPL Name Function Sequence SEQ ID No.
FLT31 itcatttctggcacagcacctTTTTTgaagttaccgtttt 320
FLT32 LE ggtgcaiiccctgcccagTTTTTctgagicaaagcat 321
FLT33 CE igatitagatctaitgtgaacagcctlTTlTctctiggaaagaaagi, 322
FLT34 tggcaatgtggteigaggagttTTTTTgaagttaccgtttt 323
FLT35 LE ggttcccctactttaagaaataatigTTTTTcigagtcaaagcat 324
FLT36 CE cagctttgcacctiatccataagTTTTTctcttggaaagaaagt 325
FLT37 LE ccgaateeatggttcacatgaaTTTTTgaagttaccgtftt 326
FLT38 LE igttttciaaticccaggtgagcTTTTTcigagtcaaagcat 327
FLT39 CE tigccctcctcgagigcttTn Tciciiggaaagaaagt 328 FLT310 LE- tgttgaaiaggtactcatctcaaagtagTTTTTgaagttaecgttii 329
FLT31 ! LE aacagaaiccgtatcatagttcigttTlTTTcigagtcaaagcat 330
FLT312 LE igccaelgatgatacaaaagcaTTTTTgaagttaccgtttt 331
FLT313 LE agtgtagiatceggtgiegtitetTTTTTcigagicaaagcai 332
FLT314 CE gggatgctttgaagaggaacaTTTTTctcttggaaagaaagi 333
FLT315 gatggtaaccaaagcigattgactTTTn gaagiiaccgiitl 334
FLT316 LE ggtagcatttaiaaaicccttticiacTTTTTctgagicaaagcat 335
FLT317 CE gtcaaittcataaieiteacttgaatiTTTTTcicttggaaagaaagt 336
FLT318 LE gacagaaaaacaaaactcttcaia tgTTTTTgaagitaccgittt 337
FLT3I9 LE aiiigtgggiaggctitaaacctT'rn rcigagtcaaagcat 338
FLT320 LE gagagaaggiccacgtacaictgTT'rrrgaagitaccgltii 339
FLT321 LE iigcicacaaggaaatgaitttcTTTTTctgagtcaaagcai 340
FLT322 LE gctgiatccgitatcaagaccciiTTTTTgaagUaccgtttt 341
FLT323 LE gcitatgaitgcaaaacltggataiTTTTTctgagtcaaagcat 342
FLT324 -LfE- ggaaiaiatattcicciggciggtTTriTgaagtiaccgiiti 343
FLT325 LE gggcatcatcatttictgcatTTTTTctgagtcaaagcat
FLT326 CE cagcgtgaacatmggtaaattTTTn ctcttggaaagaaagi 345
FLT327 CE gcacttgaggtticcttcttaiattTTTTTctctiggaaagaaagt 346
FLT328 CE itgccgatgcticlgcgaTTTTTcictiggaaagaaagi 347
Table 7
SEQ ID
FPL Name Function Sequence No. sGAPOOl CE gaamgccaigggiggaatTTTTTctcitggaaagaaagt
hsGAP002 CE ggagggatetegctcctggaTTTTTetcttggaaagaaagi 310 hsGAP003 CE ccccagcc tctccatggiTTTTTctcttggaaagaaagi 311 hsGAP004 CE gctcccccctgcaaaigagTTTTTctctiggaaagaaagt 312 hsGAP005 LE agcettgaeggtgccaigTTTTTaggcataggaceegtgtet 313 hsGAP006 LE gatgacaagcttcccgttctcTTTTTaggcaiaggaccegigtci 314 hsGAP0O7 LE agatggigatgggatttccattTTTTTaggcataggacccgtgtcl 315 hsGAPOOS LE gcaicgccccacttgaitUTTTTTaggcaiaggacccgigtci 316 hsGAP009 LE cacgacgiactcagcgccaTTTTTaggcataggacccgigtot 317 hsGAPOIO LE ggcagagatgatgaccctitigTTTlTaggc-ataggacccgigict 318 hsGAPOl l BL ggtgaagacgccagtggactc 319
Table 8
Mismatch
Number pos. from 5'
Accession ymbol Description Off-target site ^!~3'] mismatches end ef as Region
Homo sapiens
chroinosoine 6
open reading
frame 105
(C6orfl05),
transcript variant
OFF- ? NM. 00! 143948.1 C6orf105 1, mRNA UACUUUGAGACGGGUACCU 79 CDs:
Homo sapiens
major facilitator
superfamily
domain
containing 6
GFF-2 NM...017694.3 MFSD6 (MFSD6). mRNA UACCUUGAGAUGUGUACCA i 7 36 CDS;
Homo sapiens
G2/M-phase
specific E3
ubiquitin Mgase
OFF -3 'NM O S 7769.3 G2E3 (G2E3), mRNA CACUCAGAGAUAAGTJACCG I 8 14 15 39
Homo sapiens
preny [cysteine
oxidase I
(PCYOX1 ),
QFF-4 , 016297.3 PCYOX! mRNA CAUUUUAAGOUGAGUACCU 10 i3 Π 19 3UTR
Homo sapiens
pleekstrai
homology-lske
domain, family A,
member i
(PHLDA 1),
OFF-5 NM. 007350.3 PHLDA ] mRNA UACUUUUUGAGGAGUACCU 9 Yl 1 3UTR
Homo sapiens
topoisoraerase
(DNA) II binding
OFP-6 MM 007027,3 TOPBP1 $ AAUUUAAAGAUGAGUACCU 13 14 17 19 nrm.
Mismatch
os. from 5'
Accession Symbol Descri tio Off-target site [S'-S'l mismatc es end of ¾s
(TOPBP1),
m NA
Homo sapsens
myocyte enhancer
factor 2A
(MEF2A).
transcript variant
OFF-? NMJ)05587-2 MEF2A 1, naR A ACCUUUGAUGUAAGUACCU 8 !O i l IS 19 3UT
Homo sapiens
ring fmger
protein, L1M
domain
interacting
(RUM),
transcript variant
OFF-E N 01.61203 RUM AGC U AUG AGU U A AG U ACCU 8 10 15 38 19 3UTR
Homo sapiens
zinc finger,
CCHC domain
containing 6
(ZCCHC6),
transcript variant
GFF-9 NM .024617.3 ZCCHC6 AACAUGAACAUGAGUACCU I I 33 14 16 19 3UTR
Homo sapiens
z nc finger protean
148 (ZNFI 8),
OFF- 10 NM .021964.2 ZNF148 mRNA UGUUUUG A GUG AGU ACC A 1 1 7 I S MJTR
Homo sapiens
i 7
(FKBP7),
transcript variant
OFF- \ 1 NM ! S 1342.2 F BP? i, RKA GGCUUUCAGAUGAGUACAU 2 13 IS 19
Homo sapiens 5 - iiucieotidase
domain
OFF- 12 152729.2 NT5DC S containing I UACUGUGGGAUGAGUAUCU 3 32 ί ΐ 3UTR
pos. from 5'
Accession Syratiol Description Off-target site |S*-3'| end of as Region
(NTSDC 3 ),
mR A
Homo sapiens
RAB8B, member
HAS oncogene
family (RAB8B),
OFF- 13 NM 016530.2 RAB8B mRNA CACUUUAAAAUGAUUACCU 6 Π 33 19 30TR sesse
Homo sapiens
myeloid/lymphoid
or mixed-lineage
leukemia
(tdthorax
homoiog,
Drosop la);
trans located to, 6
OFF- 14 NM 0059373 MLLT6 (MLLT6). mi NA GGGUUCUCAUCUCCAAGUG 1 6 15 1 3 UTR
Homo sapiens
erythrocyte
membrane protein
band 4,1 like 4 A
(EPB41 L4A),
OFF- 15 NM 022140 EPB41 L4A mRNA CGG AACOC AUCUC AU AG UG 1 5 16 19 3UTR
Homo sapiens
glucosai sjyl (N- acety!) transferase
!, core 2
(GCNTI),
traaiscript variant
OFF- 36 NM 001097636.1 GCNTI 5, mRNA UGUCACACAUCUCAAAGUA 13 16 1 ? 19
umber
Accession Sym ol sreisiKatehes
Homo sapiens
cAMP responsive
element bfed ng
proiein 3-like 4
(CREB3L4),
QFW NM 330898 CREB3L4 mRNA UUGCAUAUAUGAGAGGGUA 3UTR
Homo sapiens
TRIO and F-aciin
binding protein
(TRJOBP),
transcript variant
OFF-2 NM 007032 3 , mRNA CUGCAUAUCUGAGCGCGCC 3UTK.
Homo sapiens
solute carrier
family 3
(gliitamate/neutrai
amino acid
transporter),
member 4
(SLC3A4).
transcript variant
OFF-3 NM 003038 SLCLA 3 , mRNA GGGAAGAUCUGAGAGCGUC ! 14 36 IS 19
Homo sapiens
DDI-ID domain
containing 1
(DDHDI),
transcript variant
OFF-4 NM 030637 DDHD i i , mRNA UUGAAUAUCUAGGAGCGUC 1 8 9 16 19 mm
Homo sapiens
RELT-like 2
(RELL2),
transcript variant
OFF-5 NM 173828 RELL2 1, mR A CUCCAUAUGAUAGAGCGUG 9 10 3 3 17 3UTR OFF-6 NM 001 193421 TSHZ2 Homo sapiens UUGGAUGGCUOGGAGCGUA % 32 33 16 3UTR
Number
Accession SymboS Descri tio Off-target site |S'-3' misKiatc e; egson
teashirt zinc
finger homeobox
2 (TSH22),
transcript variant
2, mRNA
> sapiens
ubiquitirt specific
peptidase 40 1 13 15 17 18
OF -? n USF40 (USP40), rrsRNA ACACOUGUCUGAGAGCGUC 1 3UTR
Homo sapiens
steriie alpha moiif
domain
containing 4A
(SAMD4A),
transcript variant 3 I I 13 15
OFF-8 NM 015589 SAMD4A 1, mRNA UUGAUUUUGUGAGAGCGtJA JUTR
Homo sapiens
hypoxia itiducib!e
factor 1, alpha
subunit inhibitor
(HIF 1AN),
OFF-9 NM 017902 KIFIAN mRNA GUGCAUGCCUGAGUGCGUU 6 12 13 3UT
Homo sapiens
myotubularin
related protein 3
(MTMR3),
transcript variant
OFF-10 NM 153050 1, mRNA AUGGAGAUCUOAGACCGUG 1 5 14 16
Homo sapiens
piatelet-derived
growth factor
alpha polypeptide
(PDGFA).
transcript variant
OFF- 11 NM 002607 PDGFA 3, mRNA UUGUAUACCUGAGAGCCUG \ 3 12 16 89 3IJTR
Homo sapiens
OFF- 12 NM 00 135179 ZDHHC3 zinc finger, GUGCCUAUCUGGGAGCAUG f 3 8 15 19 3UTR
Mismatch
pos. from 5'
Accession Sysnbo!l Description Off-target site |S"-3' end of as Kegioss
DHHC-type
containing 3
(ZDHHC3),
transcript variant
1 , mRNA
Homo sapiens
Rho GTPase
activating protein
1 1 A
(ARHGAPHA),
transcript variant
OFF- 13 NM. 014783 ARHGAP HA ! , mRNA GUUC A A A UUUGAG AGCGU U Π 14 17 19 5UTR sense
Homo sapiens
trop inin
associated protein
(iasiiB) (TROAP),
transcript variant
OFF-14 NM 005480 TROAP 1, mRNA UACGCUCUCAGAAACGCAC \ 5 7 13 cm
Homo sapiens
C 1GALT1- specifle
chaperone 1
(CIGALTICI),
transcript variant
OFF-15 NM .152092 CIGALTICI 1 , mRNA AGCGUUCUAAGAUAUGCAA 1 Π 15 38
Table 10
Figure imgf000078_0001
pos. from 5"
ACSeSS8£J55 Off-target site {5'-Ζ' es end of as
Homo sapiens leucine
rich repeat containing
OFF- N 0I776S.4 LRRC40 40 (LRRC40), mRNA. UGACCAAUUGACUUGAAUA 1 3 10 19
Homo sapiens SH3- domaifi GRB2-Hke
endophiKn B 3
.9 NM 016009.3 SH3GLB 1 (SH3GLB 1), mRNA GGACAAAUUUACUUCAUUA 5 IS
Homo sapiens MIDI
interacting protein ί
(gastrulstion specific
G 12 homo log
{zebraf h)) (MIDOPl }!
transcript variant 1 ,
OFF-3 021242.4 MID! l PI mRNA AAAUCAAU UUAAU UGAUUU 4 8 16 I S 19
Homo sapiens ring
finger protein 549
OFF-4 MM 173647.. RNFI49 (RNF149), rnRJNA U A ACC AGO U UUCUUGAUUA 13 18 19
Homo sapiens S AD
family member 2
(SMAD2), transcript
OFF-5 NM .005903.4 SMAD2 variiffit 1 , rnRN A UCACUAAUUUGCLIUGAUUA 1 9 15 I S 19
Homo sapiens
asparagine-Hnked
glycosylaison 10, aipha- I ,2-gkieosyhra!isferase
hornoiog B (yeast)
OFF-6 NM OQ1013620.3 ALG iOB (ALG i OB), mRNA AGACAAAUUGUGUUGAUUA 1 8 9 10 15 19
Homo sapiens ELAV
(embryonic lethal,
abnormal vision,
Drosophiia)-like S (Hu
antigen R) (ELAVL1 ),
OFF-7 ί 1419.2 ELAVL1 mRNA AG ACC AUU AA AUU U G AUUU 8 10 \ \ 13 39
um er pes. imm 5'
Accession ymbol Description Off-target site [S'-3*| es¾d of as
Homo sapiens ring
finger protein, LIM
domain interacting
i L.i M). transcrip
OFF-8 NM 056120.3 RLifvf v&nant 1, mRNA UGUCAAAIJAGACUUGAUUU EI 351719
Homo sapiens ubiqeaitin
specific peptidase 25
OFF-9 NM 0333963 USP25 (USP25), mRNA UCACA.AAUCUAAUUGAUUU 811 35 IS 9
Homo sapiens ADP- ribosyiation factor 3
OFF- SO NM 001659.2 ARF3 (ARF3), mR A AUUCCAAUUUACUGGAUUU 617 IS 39
Ηοίϊΐο sapiens down- regoiator of
transcription 1 , TBP- bsnding (negative
cofactor2) (DRI),
OFF-il NM..001938.2 DRi mRNA AGAUCAAUAUACUUGUUUA ί 411 1619
Homo sapie s
serine/arginine-rich
splicing facfor 13
(SRSFli), transcript
OFF- 12 NM Q0476 SRSF11 variant 1, mRNA AG AC A A AUCU AC U UG AUAA 12 U 1539
Homo sapiens
seitn&'ihfe !>in7t T sifie
interacting protein
(STYX), transcript
OFF- 13 Nt J4525L3 STYX variant L mRNA ACACCAAGUUACUUGCUUU 4121819 sesse
Homo sapiens post-GPi
attachment to proteins \
GFF-S4 NM.024989.3 PGAPl (PGAPl), mRNA AAAUCAAGUAAAUUGUUGA
Homo sapiens WNK
lysine deficient protein
kinase 1 (W Kt),
transcript variant 1,
OFF- 55 NM.01S979.3 WNK J mRNA UAAGUAAGUAAGUUGGUCU 1 S 151659
Table 1 1
Figure imgf000080_0001
C6orfl0525 aaaiggtgtgeaaaaettcctctTTTTTctgagtcaaagcat
80
Figure imgf000081_0001
Figure imgf000082_0001
82
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
EF2A3 tgiagjc¾gcggcattgctl Tctcttgga^gaaagj_
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001

Figure imgf000090_0001
Figure imgf000091_0001
ΚΑΒβΒΪ" ctttcctgacccgctgigtclTr'F'i' tclggaaagaaagt
91
Figure imgf000092_0001
Figure imgf000093_0001
SEQ ID
FPL Name No
EPB4IL4A20 CE ciccattttccatgtitgcagtTTTTTctctiggaaagaaagt 76S
EPB41 L4A23 CE ggciat Ltteattet ilgcitlc fXTTTTe lcttggaaagaaagt 769
EPB41L4A27 CE tg gatcacigggagaattgt'i rrri'ctcttggaaagaaagt 770
EPB41L4A1 LE caa aacaccaaccggag aaiTTTTTgaagtiaccgtitt 771
ΈΡΒ4 Ϊ Α2Π LE cccacltgctitttattc tgiaca'riTI 'Fcigagicaaagcat 772
EPB41 L4A5 LE tttcccagiactcigagtt.caaa.ttTTTTTgaagttaccgtiti 773
EPB41 L4A6 LE aaagaaigaggtttcgitacaaictT'rn'Tctgagicaaagcai 774
EPB41 L4A12 E tgcltataacgi.aiggatccaaaeTTTTTgaagitaccgtttt 775
EPB41 L4A13 LE tglcctgccactgiagcggTTTTTctgagicaaagcat 776
EPB41L4A16 I LE itcitgtcacattctgaicaggcTTTTTgaagttaccgtt 777
EPB41 L4A17 LE gciiagggiaagt.citgciicgacTTTTTcigagicaaagcat 778
EPB4IL4A18 LE gctggctgtgtitgigctattc'ri'i' ri gaagt:.accgtttt 779
EPB41L4A19 LE iaicctactgaigctgtitgaltcaTTTTTctgagtcaaagcai 780
EPB41L4A21 LE caataattttaattgttccticatral IITgaagttaccgtm 781
EPB41L4A22 EE iaaagcittttaciggtgaaggtgTTTTTcigagtcaaagcat 782
I EPB41 L4A24 LE gagatttgctictttgggtatcagTTTTTgaagttaccgtttt 783
EPB4IL4A25 LE atittcttcccacggigcafiTi'TTcigag caaagcat 784
EPB41 L4A28 LE iaagggaacittggcgacttagTTTTTgaagttaccgtitt 785
EPB41L4A29 LE g ti c ggc gacgcgtgTTTTTetgaglcaaageal 786
GCNT128 BL fctttatgggatggcatcctct 787
GCNTl 1 CE tacaaatteagge&tigalgaatTTTTTcleitggaaagaaagt 788
GCNT14 CE ccetg aaaactitggigcaaiTTTTTctctiggaaagaaagt 789
GCNTl 9 CE icttgatgaaagaagaacagteactgTTTTTetc ggaaagaaagt 790
GCNTl 10 CE ggggttcfaeaatatalttgegicTTTTTctetiggaaagaaagt 791
GCNTl 15 CE cacaigaatgcaaiagaaattctgagTTTTTctcttggaaagaaagt 792
GCNTl 18 CE aagacaltaciaaaacaggaagcgaTTTT retcitggaaagaaagt 793
GCNTl 21 CE g cagccigaacccggciTTTTTctciiggaaagaaagt 794
GCNTl 29 CE taccgcitc ccacclttelTTTTTetcitggaaagaaagt 795
GCNT12 LE gcaagcieeaagtgicigacaciTTTTTgaa.gttaccgtili 796
GCNTl 3 LE taatateaciaciaggaUci cccaTTTTTctgagicaaagcat 797
GCNTl 3 LE ta.cc iiiggatitcatltacatcaTTTT gaagttaccgltii 798
Figure imgf000095_0001
95
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
98
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
102
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
107
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
110
Figure imgf000111_0001
111
Figure imgf000112_0001
112
Figure imgf000113_0001
Figure imgf000114_0001
114
Figure imgf000115_0001
Figure imgf000116_0001
116
Figure imgf000117_0001
Figure imgf000118_0001
118
Figure imgf000119_0001
119
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
ı22
Figure imgf000123_0001

Figure imgf000124_0001
Figure imgf000125_0001
g gttggccttggttggggccttggccttggccttggtt * -· ·'·-
Figure imgf000126_0001

Claims

Claims
L A double-strarsded ribonucleic acid molecule capable of inhibiting the expression of FLT3 gene in vitro by at least 60 %, preferably by at least 70% and most preferably by at least 80%
2. The double-stranded ribonucleic acid molecule of claim !, wherein said double-stranded ribonucleic acid molecule comprises a sense strand arid an antisense strand, the antisense strand being at least partially complementary to the sense strand, whereby the sense strand comprises a sequence, which has an identity of at least 90 % to at least a portion of an mRNA encoding FLT3, wherein said sequence is (i) located its the region of complementarity of said sense strand to said aniisense strand; and (ii) wherein said sequence is less than 30 nucleotides in length,
3. The double-stranded ribonucleic acid molecule of claims 1 or 2, wherein said sense strand comprises nucleotide acid sequences depicted in SEQ ID Nos: 3, 5, 7, 1 1, 13, 21, 25 and 27 and the antisense strand is selected from the group consisting of the nucleic acid sequences depicted in SEQ ID NOs; 4, 6, S, 12, 14, 22. 26 and 28, wherein said double-stranded ribonucleic acid molecule comprises the sequence pairs selected from the group consisting of SEQ ID NOs; 3/ 4, 5/6, 7/8, 11/12, 13/14, 21/22, 25/26 and 27/21
4. The double-stranded ribonucleic acid molecule of claim 3, wherein the antisense strand f rther comprises a 3' overhang of 1-5 nucleotides in length, preferably of 1-2 nucleotides in length.
5. The double-stranded ribonucleic acid molecule of claim 4, wherein t e overhang of the antisense strand comprises uracil or nucleotides which are complementary to the rnRNA encoding FLT3.
6. The double-stranded ribonucleic acid molecule of any of claims 3 to 5, wherein the sense strand further comprises a 3' overhang of 1-5 nucleotides In length, preferably of 1-2 nucleotides in length.
7. The double-stranded ribonucleic acid molecule of claim 6 wherein the overhang of the sense strand comprises uracil or nucleotides which are identical to the niRNA encoding FLT3.
8. The double-stranded ribonucleic acid molecule of any one of claims 1 to 7, wherein said double-stranded ribonucleic acid molecule comprises at least one modified nucleotide.
9. The double-stranded ribonucleic acid molecule of claim 8, wherein said modified nucleotide is selected from the group consisting of a 2,~0-methyl modified nucleotide, a nucleotide comprising a S'-phosphorothioate group, and a terminal nucleotide linked to a choiesteryl derivative or dodecanoic acid bisdeeylamide group, a 2'-deoxy-2'-f!uoro modified nucleotide, a 2'-deoxy-modifsed nucleotide, a locked nucleotide, an abasic nucleotide, inverted deoxythymidine, I'-amino-modified nucleotide, 2!-alkyl-modif1ed nucleotide, morpholino nucleotide, a phosphoramidate, and a non-natural base comprising nucleotide.
10. The double-stranded ribonucleic acid molecule of any one of claims 8 and 9, wherein said modified nucleotide is a 2'~0~methyl modified nucleotide, a nucleotide comprising a S'-phosphorothioate group, and a deoxythymidine.
1 1. The double-stranded ribonucleic acid molecule of any one of claims 3 to 10, wherein said sense strand and / or said antisense strand comprise au overhang of 1 -2 deoxy thymidines,
12. The double-stranded ribonucleic acid molecule of any one of claims 1 to 1 1, wherein said double-stranded ribonucleic acid molecule comprises the sequence pairs selected from the group consisting of SEQ ID NOs: 149/150, 151/152, 153/154,
157/158, 159/160, 167/168, 171/172, 173/174, 293/294, 295/296 and 297/298,
13. A nucleic acid sequence encoding a sense strand and/or an antisense strand comprised in the double-stranded ribonucleic acid molecule as defined In any one of claims 1 to 12,
14. A vector comprising a regulator)' sequence operably linked to a nucleotide sequence thai encodes at least one of a sense strand or an antisense strand comprised in the double- stranded ribonucleic acid molecule as defined in any one of claims 1 to 12 or comprising the nucleic acid sequence of claim 13.
15. A cell, tissue or non-human organism comprising the double-stranded ribonucieic acid molecule as defined in any one of claims 1 to 12, the nucleic acid molecule of claim 13 or the vector of claim 14.
16. A pharmaceutical composition comprising the double-stranded ribonucleic acid molecule as defined in any one of claims 1 to 12, the nucleic acid molecule of claim 13, the vector of claim 14 or the cell or tissue of claim! 5.
17. The pharmaceutical composition of claim 16, further comprising a pharmaceutically acceptable carrier, stablilizer and/or diluent.
18. A method for inhibiting the expression of FLT3 gene in a cell, a tissue or an organism comprising the following steps:
(a) introducing into the cell, tissue or organism the double-stranded ribonucleic acid molecule as defined in any one of claims 1 to 12, the nucleic acid molecule of claim 13», the vector of claim 14; and
(b) maintaining the cell, tissue or organism produced irs step (a) for a time sufficient to obtain degradation of the mRNA transcript of a FLT3 gene, thereby inhibiting expression of a FLT3 gene in the cell.
19. A method of treating, preventing or managing pathological conditions and diseases caused by the expression of the FLT3 gene disease comprising administering to a subject in need of such treatment, prevention or management a therapeutically or prophylacticaiiy effective amount of a the double-stranded ribonucleic acid molecule as defined in any one of claims 1 to 12, a nucleic acid molecule of claim 13, a vector of claim 14 and/or a pharmaceutical composition as defined in claims 16 or 18.
20. The method of claim 1 , wherein said subject is a human.
21. A double-stranded ribonucleic acid molecule as defined in any one of claims ί to 12, a nucleic acid molecule of claim 13, a vector of claim 14 and/or a pharmaceutical composition as defined in claims 16 or IS for use in treating inflammation and proliferative disorders.
22. Use of a the do ble-straiided ribonucleic acid molecule as defined in any one of claims 1 to 12, a nucleic acid molecule of claim 13, a vector of claim 14 aind/or a cell or tissue of claim 15 for the preparation of a pharmaceutical composition for the treatment of inflammation and proliferative disorders*
PCT/US2011/064896 2010-12-17 2011-12-14 Compositions and methods for inhibiting expression of flt3 genes WO2012094115A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EPEP10195761 2010-12-17
EP10195761 2010-12-17

Publications (1)

Publication Number Publication Date
WO2012094115A1 true WO2012094115A1 (en) 2012-07-12

Family

ID=46457659

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/064896 WO2012094115A1 (en) 2010-12-17 2011-12-14 Compositions and methods for inhibiting expression of flt3 genes

Country Status (1)

Country Link
WO (1) WO2012094115A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016149020A1 (en) * 2015-03-17 2016-09-22 Arrowhead Research Corporation Rna interference agents
WO2018005608A1 (en) * 2016-06-28 2018-01-04 Northeast Ohio Medical University Compositions and methods for preventing bone loss and/or stimulating bone healing
US10982015B2 (en) * 2018-07-06 2021-04-20 Zhejiang Gongshang University PEG-ACS/M-siRNA nanocomposite, application thereof, and method for reducing histamine content during fishmeal storage
US11015198B2 (en) 2017-04-05 2021-05-25 Silence Therapeutics Gmbh Products and compositions

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005035004A1 (en) * 2003-10-09 2005-04-21 Takara Bio Inc. COMPOSITION FOR INHIBITING FUNCTION OF HUMAN Flt3
US20100291681A1 (en) * 2002-11-14 2010-11-18 Dharmacon, Inc. Methods and compositions for selecting siRNA of improved functionality

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100291681A1 (en) * 2002-11-14 2010-11-18 Dharmacon, Inc. Methods and compositions for selecting siRNA of improved functionality
WO2005035004A1 (en) * 2003-10-09 2005-04-21 Takara Bio Inc. COMPOSITION FOR INHIBITING FUNCTION OF HUMAN Flt3

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE EMBL [online] 31 March 2012 (2012-03-31), KATO ET AL., retrieved from http://www.ebi.ac.uklfools/dbfetch/emblfetch?style=html8id=dd152595 Database accession no. DD152595 *
DATABASE EMBL [online] 31 March 2012 (2012-03-31), KATO ET AL., retrieved from http://www.ebi.ac.uklToolsldbfetch/emblfetch?style=html&id=dd152596 Database accession no. DD152596 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016149020A1 (en) * 2015-03-17 2016-09-22 Arrowhead Research Corporation Rna interference agents
WO2018005608A1 (en) * 2016-06-28 2018-01-04 Northeast Ohio Medical University Compositions and methods for preventing bone loss and/or stimulating bone healing
US11015198B2 (en) 2017-04-05 2021-05-25 Silence Therapeutics Gmbh Products and compositions
US10982015B2 (en) * 2018-07-06 2021-04-20 Zhejiang Gongshang University PEG-ACS/M-siRNA nanocomposite, application thereof, and method for reducing histamine content during fishmeal storage

Similar Documents

Publication Publication Date Title
KR102554783B1 (en) Compositions and Methods for Inhibiting Gene Expression of Hepatitis B Virus
US20110119781A1 (en) Compositions and Methods for Inhibiting Expression of TGF-BETA Receptor Genes
WO2012094115A1 (en) Compositions and methods for inhibiting expression of flt3 genes
US11920134B2 (en) Compositions and methods for inhibiting expression of RRM2 genes
US20110112176A1 (en) Compositions and methods for inhibiting expression of kif10 genes
WO2011073218A1 (en) Compositons and methods for inhibiting expression of il-18 genes
US20100197773A1 (en) Compositions and methods for inhibiting expression of ptp1b genes
WO2012082894A1 (en) Compositions and methods for inhibiting expression of mll genes
AU2010247389A1 (en) Compositions and methods for inhibiting expression of glucocorticoid receptor (GCR) genes
WO2011095495A1 (en) Compositions and methods for inhibiting expression of ikk2 genes

Legal Events

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

Ref document number: 11854639

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11854639

Country of ref document: EP

Kind code of ref document: A1