Compositions and methods for inhibiting expression of Eg5 gene

Information

  • Patent Grant
  • 7718629
  • Patent Number
    7,718,629
  • Date Filed
    Friday, March 30, 2007
    17 years ago
  • Date Issued
    Tuesday, May 18, 2010
    14 years ago
Abstract
The invention relates to a double-stranded ribonucleic acid (dsRNA) for inhibiting the expression of the Eg5 gene (Eg5 gene), comprising an antisense strand having a nucleotide sequence which is less that 30 nucleotides in length, generally 19-25 nucleotides in length, and which is substantially complementary to at least a part of the Eg5 gene. The invention also relates to a pharmaceutical composition comprising the dsRNA together with a pharmaceutically acceptable carrier; methods for treating diseases caused by Eg5 expression and the expression of the Eg5 gene using the pharmaceutical composition; and methods for inhibiting the expression of the Eg5 gene in a cell.
Description
FIELD OF THE INVENTION

This invention relates to double-stranded ribonucleic acid (dsRNA), and its use in mediating RNA interference to inhibit the expression of the Eg5 gene and the use of the dsRNA to treat pathological processes mediated by Eg5 expression, such as cancer, alone or in combination with a dsRNA targeting vacular endothelian growth factor (VEGF).


BACKGROUND OF THE INVENTION

The maintenance of cell populations within an organism is governed by the cellular processes of cell division and programmed cell death. Within normal cells, the cellular events associated with the initiation and completion of each process is highly regulated. In proliferative disease such as cancer, one or both of these processes may be perturbed. For example, a cancer cell may have lost its regulation (checkpoint control) of the cell division cycle through either the overexpression of a positive regulator or the loss of a negative regulator, perhaps by mutation.


Alternatively, a cancer cell may have lost the ability to undergo programmed cell death through the overexpression of a negative regulator. Hence, there is a need to develop new chemotherapeutic drugs that will restore the processes of checkpoint control and programmed cell death to cancerous cells.


One approach to the treatment of human cancers is to target a protein that is essential for cell cycle progression. In order for the cell cycle to proceed from one phase to the next, certain prerequisite events must be completed. There are checkpoints within the cell cycle that enforce the proper order of events and phases. One such checkpoint is the spindle checkpoint that occurs during the metaphase stage of mitosis. Small molecules that target proteins with essential functions in mitosis may initiate the spindle checkpoint to arrest cells in mitosis. Of the small molecules that arrest cells in mitosis, those which display anti-tumor activity in the clinic also include apoptosis, the morphological changes associated with programmed cell death. An effective chemotherapeutic for the treatment of cancer may thus be one which induces checkpoint control and programmed cell death. Unfortunately, there are few compounds available for controlling these processes within the cell. Most compounds known to cause mitotic arrest and apoptosis act as tubulin binding agents. These compounds alter the dynamic instability of microtubules and indirectly alter the function/structure of the mitotic spindle thereby causing mitotic arrest. Because most of these compounds specifically target the tubulin protein which is a component of all microtubules, they may also affect one or more of the numerous normal cellular processes in which microtubules, they may also affect one or more of the numerous normal cellular processes in which microtubules have a role. Hence, there is also a need for small molecules that more specifically target proteins associated with proliferating cells.


Eg5 is one of several kinesin-like motor proteins that are localized to the mitotic spindle and known to be required for formation and/or function of the bipolar mitotic spindle. Recently, there was a report of a small molecule that disturbs bipolarity of the mitotic spindle (Mayer, T. U. et. al. 1999. Science 286(5441) 971-4, herein incorporated by reference). More specifically, the small molecule induced the formation of an aberrant mitotic spindle wherein a monoastral array of microtubules emanated from a central pair of centrosomes, with chromosomes attached to the distal ends of the microtubules. The small molecule was dubbed “monastrol” after the monoastral array. This monoastral array phenotype had been previously observed in mitotic cells that were immunodepleted of the Eg5 motor protein. This distinctive monoastral array phenotype facilitated identification of monastrol as a potential inhibitor of Eg5. Indeed, monastrol was further shown to inhibit the Eg5 motor-driven motility of microtubules in an in vitro assay. The Eg5 inhibitor monastrol had no apparent effect upon the related kinesin motor or upon the motor(s) responsible for golgi apparatus movement within the cell. Cells that display the monoastral array phenotype either through immunodepletion of Eg5 or monastrol inhibition of Eg5 arrest in M-phase of the cell cycle. However, the mitotic arrest induced by either immunodepletion or inhibition of Eg5 is transient (Kapoor, T. N., 2000, J Cell Biol 150(5) 975-80). Both the monoastral array phenotype and the cell cycle arrest in mitosis induced by monastrol are reversible. Cells recover to form a normal bipolar mitotic spindle, to complete mitosis and to proceed through the cell cycle and normal cell proliferation. These data suggest that a small molecule inhibitor of Eg5 which induced a transient mitotic arrest may not be effective for the treatment of cancer cell proliferation. Nonetheless, the discovery that monastrol causes mitotic arrest is intriguing and hence there is a need to further study and identify compounds which can be used to modulate the Eg5 motor protein in a manner that would be effective in the treatment of human cancers. There is also a need to explore the use of these compounds in combination with other antineoplastic agents.


VEGF (also known as vascular permeability factor, VPF) is a multifunctional cytokine that stimulates angiogenesis, epithelial cell proliferation, and endothelial cell survival. VEGF can be produced by a wide variety of tissues, and its overexpression or aberrant expression can result in a variety disorders, including cancers and retinal disorders such as age-related macular degeneration and other angiogenic disorders.


Recently, double-stranded RNA molecules (dsRNA) have been shown to block gene expression in a highly conserved regulatory mechanism known as RNA interference (RNAi)). WO 99/32619 (Fire et al.) discloses the use of a dsRNA of at least 25 nucleotides in length to inhibit the expression of genes in C. elegans. dsRNA has also been shown to degrade target RNA in other organisms, including plants (see, e.g., WO 99/53050, Waterhouse et al., and WO 99/61631, Heifetz et al.), Drosophila (see e.g., Yang, D., et al., Curr. Biol. (2000) 10:1191-1200), and mammals (see WO 00/44895, Limmer; and DE 101 00 586.5, Kreutzer et al.). This natural mechanism has now become the focus for the development of a new class of pharmaceutical agents for treating disorders that are caused by the aberrant or unwanted regulation of a gene.


Despite significant advances in the field of RNAi and advances in the treatment of pathological processes mediated by Eg5 expression, there remains a need for an agent that can selectively and efficiently silence the Eg5 gene using the cell's own RNAi machinery that has both high biological activity and in vivo stability, and that can effectively inhibit expression of a target Eg5 gene for use in treating pathological processes mediated by Eg5 expression.


SUMMARY OF THE INVENTION

The invention provides double-stranded ribonucleic acid (dsRNA), as well as compositions and methods for inhibiting the expression of the Eg5 gene in a cell or mammal using such dsRNA, alone or in combination with a dsRNA targeting VEGF. The invention also provides compositions and methods for treating pathological conditions and diseases caused by the expression of the Eg5 gene, such as in cancer. The dsRNA of the invention comprises an RNA strand (the antisense strand) having a region which is less than 30 nucleotides in length, generally 19-24 nucleotides in length, and is substantially complementary to at least part of an mRNA transcript of the Eg5 gene.


In one embodiment, the invention provides double-stranded ribonucleic acid (dsRNA) molecules for inhibiting the expression of the Eg5 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. The antisense strand comprises a nucleotide sequence which is substantially complementary to at least part of an mRNA encoding Eg5, and the region of complementarity is less than 30 nucleotides in length, generally 19-24 nucleotides in length. The dsRNA, upon contacting with a cell expressing the Eg5, inhibits the expression of the Eg5 gene by at least 40%.


For example, the dsRNA molecules of the invention can be comprised of a first sequence of the dsRNA that is selected from the group consisting of the sense sequences of Tables 1-3 and the second sequence is selected from the group consisting of the antisense sequences of Tables 1-3. The dsRNA molecules of the invention can be comprised of naturally occurring nucleotides or can be comprised of at least one modified nucleotide, such as a 2′-O-methyl modified nucleotide, a nucleotide comprising a 5′-phosphorothioate group, and a terminal nucleotide linked to a cholesteryl derivative. Alternatively, the modified nucleotide may be chosen from the group of: a 2′-deoxy-2′-fluoro modified nucleotide, a 2′-deoxy-modified nucleotide, a locked nucleotide, an abasic nucleotide, 2′-amino-modified nucleotide, 2′-alkyl-modified nucleotide, morpholino nucleotide, a phosphoramidate, and a non-natural base comprising nucleotide. Generally, such modified sequence will be based on a first sequence of said dsRNA selected from the group consisting of the sense sequences of Tables 1-3 and a second sequence selected from the group consisting of the antisense sequences of Tables 1-3.


In another embodiment, the invention provides a cell comprising one of the dsRNAs of the invention. The cell is generally a mammalian cell, such as a human cell.


In another embodiment, the invention provides a pharmaceutical composition for inhibiting the expression of the Eg5 gene in an organism, generally a human subject, comprising one or more of the dsRNA of the invention and a pharmaceutically acceptable carrier or delivery vehicle.


In another embodiment, the invention provides a method for inhibiting the expression of the Eg5 gene in a cell, comprising the following steps:

    • (a) introducing into the cell a double-stranded ribonucleic acid (dsRNA), wherein 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. The antisense strand comprises a region of complementarity which is substantially complementary to at least a part of a mRNA encoding Eg5, and wherein the region of complementarity is less than 30 nucleotides in length, generally 19-24 nucleotides in length, and wherein the dsRNA, upon contact with a cell expressing the Eg5, inhibits expression of the Eg5 gene by at least 40%; and
    • (b) maintaining the cell produced in step (a) for a time sufficient to obtain degradation of the mRNA transcript of the Eg5 gene, thereby inhibiting expression of the Eg5 gene in the cell.


In another embodiment, the invention provides methods for treating, preventing or managing pathological processes mediated by Eg5 expression, e.g. cancer, comprising administering to a patient in need of such treatment, prevention or management a therapeutically or prophylactically effective amount of one or more of the dsRNAs of the invention.


In another embodiment, the invention provides vectors for inhibiting the expression of the Eg5 gene in a cell, comprising a regulatory sequence operable linked to a nucleotide sequence that encodes at least one strand of one of the dsRNA of the invention.


In another embodiment, the invention provides a cell comprising a vector for inhibiting the expression of the Eg5 gene in a cell. The vector comprises a regulatory sequence operably linked to a nucleotide sequence that encodes at least one strand of one of the dsRNA of the invention.


In a further embodiment, the invention provides the Eg5 dsRNA and the uses thereof as described above in combination with a second dsRNA targeting the VEGF mRNA. A combination of a dsRNA targeting Eg5 and a second dsRNA targeting VEGF provides complementary and synergiatic activity for treating hyperproliferative discords, particulary hepatic carcinoma.





BRIEF DESCRIPTION OF THE FIGURES

No Figures are presented.





DETAILED DESCRIPTION OF THE INVENTION

The invention provides double-stranded ribonucleic acid (dsRNA), as well as compositions and methods for inhibiting the expression of the Eg5 gene in a cell or mammal using the dsRNA. The invention also provides compositions and methods for treating pathological conditions and diseases in a mammal caused by the expression of the Eg5 gene using dsRNA. dsRNA directs the sequence-specific degradation of mRNA through a process known as RNA interference (RNAi). The invention further provides this dsRNA in combination with a second dsRNA that inhibits the expression of the VEGF gene.


The dsRNAs of the invention comprises an RNA strand (the antisense strand) having a region which is less than 30 nucleotides in length, generally 19-24 nucleotides in length, and is substantially complementary to at least part of an mRNA transcript of the Eg5 gene. The use of these dsRNAs enables the targeted degradation of mRNAs of genes that are implicated in replication and or maintenance of cancer cells in mammals. Using cell-based and animal assays, the present invention have demonstrated that very low dosages of these dsRNA can specifically and efficiently mediate RNAi, resulting in significant inhibition of expression of the Eg5 gene. Thus, the methods and compositions of the invention comprising these dsRNAs are useful for treating pathological processes mediated by EG5 expression, e.g. cancer, by targeting a gene involved in mitotic division.


The following detailed description discloses how to make and use the dsRNA and compositions containing dsRNA to inhibit the expression of the Eg5 gene, as well as compositions and methods for treating diseases and disorders caused by the expression of Eg5, such as cancer, alone or in combination with a second dsRNA targeting the VEGF gene. The pharmaceutical compositions of the invention comprise a dsRNA having an antisense strand comprising a region of complementarity which is less than 30 nucleotides in length, generally 19-24 nucleotides in length, and is substantially complementary to at least part of an RNA transcript of the Eg5 gene, together with a pharmaceutically acceptable carrier. As discussed above, such compositions can further include a second dsRNA targeting VEGF.


Accordingly, certain aspects of the invention provide pharmaceutical compositions comprising the dsRNA of the invention together with a pharmaceutically acceptable carrier, methods of using the compositions to inhibit expression of the Eg5 gene, and methods of using the pharmaceutical compositions to treat diseases caused by expression of the Eg5 gene. The invention further provides the above pharmaceutical compositions further containing a second dsRNA designed to inhibit the expression of VEGF.


I. 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” and “U” each generally stand for a nucleotide that contains guanine, cytosine, adenine, and uracil as a base, respectively. However, it will be understood that the term “ribonucleotide” or “nucleotide” can also refer to a modified nucleotide, as further detailed below, or a surrogate replacement moiety. The skilled person is well aware that guanine, cytosine, adenine, and uracil may be replaced by other moieties without substantially altering the base pairing properties of an oligonucleotide comprising a nucleotide bearing such replacement moiety. For example, without limitation, a nucleotide comprising inosine as its base may base pair with nucleotides containing adenine, cytosine, or uracil. Hence, nucleotides containing uracil, guanine, or adenine may be replaced in the nucleotide sequences of the invention by a nucleotide containing, for example, inosine. Sequences comprising such replacement moieties are embodiments of the invention.


As used herein, “Eg5” refers to the human kinesine family member 11, which is also known in KIF11, Eg5, KNSL1 or TRIP5. Eg5 sequence can be found as NCBI GeneID:3832, HGNC ID: HGNC:6388 and RefSeq ID number NM004523.


As used herein, “target sequence” refers to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during the transcription of the Eg5 gene, including mRNA that is a product of RNA processing of a primary transcription product.


As used hereing, VEGF, also known as vascular permeability factor, is an angiogenic growth factor. VEGF is a homodimeric 45 kDa glycoprotein that exists in at least three different isoforms. VEGF isoforms are expressed in endothelial cells. The VEGF gene contains 8 exons that express a 189-amino acid portein isoform. A 165-amino acid isoform lacks the residues encoded by exon 6, whereas a 121-amino acid isoform lacks the residues encoded by exons 6 and 7, VEGF145 is an isoform predicted to contain 145 amino acids and to lack exon 7. VEGF can act on endothelial cells by binding to an endothelial tyrosine kinase receptor, such as Flt-1 (VEGFR-1) or KDR/flk-1 (VEGFR-2). VEGFR-2 is expressed in endothelial cells and is involved in endothelial cell differentiation and vasculogenesis. A third receptor, VEGFR-3 has been implicated in lymphogenesis.


The various isoforms have different biologic activities and clinical implications. For example, VEGF145 includes angiogenesis and like VEGF189 (but unlike VEGF165) VEGF145 binds efficiently to the extracellular matrix by a mechanism that is not dependent on extracellular matrix-associated heparin sulfates. VEGF displays activity as an endothelial cell mitogen and chemoattractant in vitro and induces vascular permeability and angiogenesis in vivo. VEGF is secreted by a wide variety of cancer cell types and promotes the growth of tumors by inducing the development of tumor-associated vasculature. Inhibition of VEGF function has been shown to limit both the growth of primary experimental tumors as well as the incidence of metastases in immunocompromised mice. Various dsRNAs directed to VEGF are described in co-pending U.S. Ser. Nos. 11/078,073 and 11/340,080, herein incorporated by reference).


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.


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, as will be understood by the skilled person. Such conditions can, for example, be stringent conditions, where stringent conditions may include: 400 mM NaCl, 40 mM PIPES pH 6.4, 1 mM EDTA, 50° C. or 70° C. for 12-16 hours followed by washing. Other conditions, such as physiologically relevant conditions as may be encountered inside an organism, can apply. The skilled person will be able to determine the set of conditions most appropriate for a test of complementarity of two sequences in accordance with the ultimate application of the hybridized nucleotides.


This includes 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. Such sequences can be referred to as “fully complementary” with respect to each other herein. However, where a first sequence is referred to as “substantially complementary” with respect to a second sequence herein, the two-sequences can be fully complementary, or they may form one or more, but generally not more than 4, 3 or 2 mismatched base pairs upon hybridization, while retaining the ability to hybridize under the conditions most relevant to their ultimate application. However, where two oligonucleotides are designed to form, upon hybridization, one or more single stranded overhangs, such overhangs shall not be regarded as mismatches with regard to the determination of complementarity. For example, a dsRNA comprising one oligonucleotide 21 nucleotides in length and another oligonucleotide 23 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 21 nucleotides that is fully complementary to the shorter oligonucleotide, may yet be referred to as “fully complementary” for the purposes of the invention.


“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 far as the above requirements with respect to their ability to hybridize are fulfilled.


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 dsRNA, or between the antisense strand of a dsRNA and a target sequence, as will be understood from the context of their use.


As used herein, a polynucleotide which is “substantially complementary to at least part of” a messenger RNA (mRNA) refers to a polynucleotide which is substantially complementary to a contiguous portion of the mRNA of interest (e.g., encoding Eg5). For example, a polynucleotide is complementary to at least a part of a Eg5 mRNA if the sequence is substantially complementary to a non-interrupted portion of a mRNA encoding Eg5.


The term “double-stranded RNA” or “dsRNA”, as used herein, refers to a complex of ribonucleic acid molecules, having a duplex structure comprising two anti-parallel and substantially complementary, as defined above, nucleic acid strands,. The two strands forming the duplex structure may be 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 covalently 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. The maximum number of base pairs is the number of nucleotides in the shortest strand of the dsRNA minus any overhangs that are present in the duplex. In addition to the duplex structure, a dsRNA may comprise one or more nucleotide overhangs.


As used herein, 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. “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” dsRNA 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, as defined herein. Where the region of complementarity is not fully complementary to the target sequence, the mismatches are most tolerated in the terminal regions and, if present, are generally in a terminal region or regions, e.g., within 6, 5, 4, 3, or 2 nucleotides of the 5′ and/or 3′ terminus.


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.


“Introducing into a cell”, when referring to a dsRNA, means facilitating uptake or absorption into the cell, as in 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 cell 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, dsRNA can be injected into a tissue site or administered systemically. In vitro introduction into a cell includes methods known in the art such as electroporation and lipofection.


The terms “silence” and “inhibit the expression of”, in as far as they refer to the Eg5 gene, herein refer to the at least partial suppression of the expression of the Eg5 gene, as manifested by a reduction of the amount of mRNA transcribed from the Eg5 gene which may be isolated from a first cell or group of cells in which the Eg5 gene is transcribed and which has or have been treated such that the expression of the Eg5 gene is inhibited, as compared to a second cell or group of cells substantially identical to the first cell or group of cells but which has or have not been so treated (control cells). The degree of inhibition is usually expressed in terms of










(

mRNA





in





control





cells

)

-

(

mRNA





in





treated





cells

)



(

mRNA





in





control





cells

)


·
100


%




Alternatively, the degree of inhibition may be given in terms of a reduction of a parameter that is functionally linked to Eg5 gene transcription, e.g. the amount of protein encoded by the Eg5 gene which is secreted by a cell, or the number of cells displaying a certain phenotype, e.g. apoptosis. In principle, Eg5 gene silencing may be determined in any cell expressing the target, either constitutively or by genomic engineering, and by any appropriate assay. However, when a reference is needed in order to determine whether a given dsRNA inhibits the expression of the Eg5 gene by a certain degree and therefore is encompassed by the instant invention, the assay provided in the Examples below shall serve as such reference.


For example, in certain instances, expression of the Eg5 gene (or VEGF gene) is suppressed by at least about 20%, 25%, 35%, or 50% by administration of the double-stranded oligonucleotide of the invention. In some embodiment, the Eg5 gene is suppressed by at least about 60%, 70%, or 80% by administration of the double-stranded oligonucleotide of the invention. In some embodiments, the Eg5 gene is suppressed by at least about 85%, 90%, or 95% by administration of the double-stranded oligonucleotide of the invention. Tables 1-3 provides values for inhibition of expression using various Eg5 dsRNA molecules at various concentrations.


As used herein in the context of Eg5 expression, the terms “treat”, “treatment”, and the like, refer to relief from or alleviation of pathological processes mediated by Eg5 expression. In the context of the present invention insofar as it relates to any of the other conditions recited herein below (other than pathological processes mediated by Eg5 expression), the terms “treat”, “treatment”, and the like mean to relieve or alleviate at least one symptom associated with such condition, or to slow or reverse the progression of such condition, such as the slowing and progression of hepatic carcinoma.


As used herein, the phrases “therapeutically effective amount” and “prophylactically effective amount” refer to an amount that provides a therapeutic benefit in the treatment, prevention, or management of pathological processes mediated by Eg5 expression or an overt symptom of pathological processes mediated by Eg5 expression (alone or in combination with VEGF expression). The specific amount that is therapeutically effective can be readily determined by ordinary medical practitioner, and may vary depending on factors known in the art, such as, e.g. the type of pathological processes mediated by Eg5 expression, the patient's history and age, the stage of pathological processes mediated by Eg5 expression, and the administration of other anti-pathological processes mediated by Eg5 expression agents.


As used herein, a “pharmaceutical composition” comprises a pharmacologically effective amount of a dsRNA and a pharmaceutically acceptable carrier. As used herein, “pharmacologically effective amount,” “therapeutically effective amount” or simply “effective amount” refers to that amount of an RNA effective to produce the intended pharmacological, therapeutic or preventive result. For example, if a given clinical treatment is considered effective when there is at least a 25% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 25% reduction in that parameter.


The term “pharmaceutically acceptable carrier” refers to a carrier for administration of a therapeutic agent. Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, 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. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.


As used herein, a “transformed cell” is a cell into which a vector has been introduced from which a dsRNA molecule may be expressed.


II. Double-stranded Ribonucleic Acid (dsRNA)


In one embodiment, the invention provides double-stranded ribonucleic acid (dsRNA) molecules for inhibiting the expression of the Eg5 gene (alone or incombination with a second dsRNA for inhibiting the expression of VEGF) in a cell or mammal, wherein the dsRNA comprises an antisense strand comprising a region of complementarity which is complementary to at least a part of an mRNA formed in the expression of the Eg5 gene, and wherein the region of complementarity is less than 30 nucleotides in length, generally 19-24 nucleotides in length, and wherein said dsRNA, upon contact with a cell expressing said Eg5 gene, inhibits the expression of said Eg5 gene by at least 40%. The dsRNA comprises two RNA strands that are sufficiently complementary to hybridize to form a duplex structure. One strand of the dsRNA (the antisense strand) comprises a region of complementarity that is substantially complementary, and generally fully complementary, to a target sequence, derived from the sequence of an mRNA formed during the expression of the Eg5 gene, the other strand (the sense strand) comprises a region which is complementary to the antisense strand, such that the two strands hybridize and form a duplex structure when combined under suitable conditions. Generally, the duplex structure us between 15 and 30, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 base pairs in length. Similarly, the region of complementarity to the target sequence is between 15 and 30, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 nucleotides in length. The dsRNA of the invention may further comprise one or more single-stranded nucleotide overhang(s). The dsRNA can be synthesized by stranded methods known in the art as further discussed below, e.g., by use of an automated DNA synthesizer, such as are commercially available from, for example, Biosearch, Applied Biosystems, Inc. In a preferred embodiment, the Eg5 gene is the human Eg5 gene. In specific embodiments, the antisense strand of the dsRNA comprises the sense sequences of Tables 1-3 and the second sequence is selected from the group consisting of the antisense sequences of Tables 1-3. Alternative antisense agents that target elsewhere in the target sequence provided in Tables 1-3 can readily be determined using the target sequence and the flanking Eg5 sequence. In embodiments using a second dsRNA targeting VEGF, such agents are exemplified in the Examples and in co-pending U.S. Ser. Nos. 11/078,073 and 11/340,080, herein incorporated by reference.


The dsRNA will comprise at least two nucleotide sequence selected from the groups of sequences provided in Tables 1-3. One of the two sequences is complementary to the other of the two sequences, with one of the sequences being substantially complementary to a sequence of an mRNA generated in the expression of the Eg5 gene. As such, the dsRNA will comprises two oligonucleotides, wherein one oligonucleotide is described as the sense strand in Tables 1-3 and the second oligonucleotide is described as the antisense strand in Tables 1-3.


The skilled person is well aware that dsRNAs comprising a duplex structure of between 20 and 23, but specifically 21, base pairs have been hailed as particularly effective in inducing RNA interference (Elbashir et al., EMBO 2001, 20:6877-6888). However, others have found that shorter or longer dsRNAs can be effective as well. In the embodiments described above, by virtue of the nature of the oligonucleotide sequences provided in Tables 1-3, the dsRNAs of the invention can comprise at least one strand of a length of minimally 21 nt. It can be reasonably expected that shorter dsRNAs comprising one of the sequences of Tables 1-3 minus only a few nucleotides on one or both ends may be similarly effective as compared to the dsRNAs described above. Hence, dsRNAs comprising a partial sequence of at least 15, 16, 17, 18, 19, 20, or more contiguous nucleotides from one of the sequences of Tables 1-3, and differing in their ability to inhibit the expression of the Eg5 gene in a FACS assay as described herein below by not more than 5, 10, 15, 20, 25, or 30% inhibition from a dsRNA comprising the full sequence, are contemplated by the invention. Further dsRNAs that cleave within the target sequence provided in Tables 1-3 can readily be made using the Eg5 sequence and the target sequence provided.


In addition, the RNAi agents provided in Tables 1-3 identify a site in the Eg5 mRNA that is susceptible to RNAi based cleavage. As such the present invention further includes RNAi agents that target within the sequence targeted by one of the agents of the present invention. As used herein a second RNAi agent is said to target within the sequence of a first RNAi agent if the second RNAi agent cleaves the message anywhere within the mRNA that is complementary to the antisense strand of the first RNAi agent. Such a second agent will generally consist of at least 15 contiguous nucleotides from one of the sequences provided in Tables 1-3 coupled to additional nucleotide sequences taken from the region contiguous to the selected sequence in the Eg5 gene. For example, the last 15 nucleotides of SEQ ID NO:1 combined with the next 6 nucleotides from the target Eg5 gene produces a single strand agent of 21 nucleotides that is based on one of the sequences provided in Tables 1-3.


The dsRNA of the invention can contain one or more mismatches to the target sequence. In a preferred embodiment, the dsRNA of the invention contains no more than 3 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 in the center of the region of complementarity. If the antisense strand of the dsRNA contains mismatches to the target sequence, it is preferable that the mismatch be restricted to 5 nucleotides from either end, for example 5, 4, 3, 2, or 1 nucleotide from either the 5′ or 3′ end of the region of complementarity. For example, for a 23 nucleotide dsRNA strand which is complementary to a region of the Eg5 gene, the dsRNA generally does not contain any mismatch within the central 13 nucleotides. The methods described within the invention can be used to determine whether a dsRNA containing a mismatch to a target sequence is effective in inhibiting the expression of the Eg5 gene. Consideration of the efficacy of dsRNAs with mismatches in inhibiting expression of the Eg5 gene is important, especially if the particular region of complementarity in the Eg5 gene is known to have polymorphic sequence variation within the population.


In one embodiment, at least one end of the dsRNA has a single-stranded nucleotide overhang of 1 to 4, generally 1 or 2 nucleotides. dsRNAs 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. Generally, the single-stranded overhang is located at the 3′-terminal end of the antisense strand or, alternatively, at the 3′-terminal end of the sense strand. The dsRNA may also have a blunt end, generally located at the 5′-end of the antisense strand. Such dsRNAs have improved stability and inhibitory activity, thus allowing administration at low dosages, e.g., less than 5 mg/kg body weight of the recipient per day. Generally, 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.


In yet another embodiment, the dsRNA is 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”, Beaucage, S. L. et al. (Edrs.), John Wiley & Sons, Ind., New York, N.Y., USA, which is hereby incorporated herein by reference. Specific examples of preferred dsRNA compounds useful in this invention include dsRNAs containing modified backbones or no natural internucleoside linkages. As defined in this specification, dsRNAs having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified dsRNAs that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides.


Preferred modified dsRNA backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3′-5′ linkages, 2′-5′ 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 U.S. patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,195; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,316; 5,550,111; 5,563,253; 5,571,799; 5,587,361; and 5,625,050, each of which in herein incorporated by reference.


Preferred modified dsRNA backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloakyl internucleoside linkages, mixed heteroatoms and alkyl or cycloalkyl internucleoside linkages, or ore or more short chain heteroatomic or heterocyclic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts.


Representative U.S. patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,64,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; and, 5,677,439, each of which is herein incorporated by reference.


In other preferred dsRNA mimetics, both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups. The base units are maintained for hybridization with an appropriate nucleic acid target compound. One such oligomeric compound, and dsRNA mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA). In PNA compounds, the sugar backbone of an dsRNA is replaced with an amide containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative U.S. patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262; each of which is herein incorporated by reference. Further teaching of PNA compounds can be found in Nielsen et al., Science, 1991, 254, 1497-1500.


Most preferred embodiments of the invention are dsRNAs with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular —CH.sub.2-NH—CH.sub.2-, —CH.sub.2-N(CH.sub.3)-O—CH.sub.2-[known as a methylene (methylimino) or MMI backbone], —CH.sub.2-O—N(CH.sub.3)-CH.sub.2-, —CH.sub.2-N(CH.sub.3)-N(CH.sub.3)—CH.sub.2- and -N(CH.sub.3)-CH.sub.2-CH.sub.2-[wherein the native phosphodiester backbone is represented as —O—P—O—CH.sub.2-] of the above-referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above-referenced U.S. Pat. No. 5,602,240. Also preferred are dsRNAs having morepholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506.


Modified dsRNAs may also contain one or more substituted sugar moieties. Preferred dsRNAs comprise one of the following at the 2′ position: OH; F; O—, S—, or N-alkyl; O—, S—, or N-alkenyl; O—, S— or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C.sub.1 to C.sub.10 alkyl or C.sub.2 to C.sub.10 alkenyl and alkynyl. Particularly preferred are O[(CH.sub.2).sub.nO].sub.mCH.sub.3, (CH.sub.2).ub.nOCH.sub.3, O(CH.sub.2).sub.nNH.sub.2, O(CH.sub.2).xub.nCH.sub.3, O(CH.sub.2).sub.nONH.sub.2, and O(CH.sub.2).sub.nON[(CH.sub.2).sub.nCH.su-b.3)].sub.2, where n and m are from 1 to about 10. Other preferred dsRNAs comprise one of the following at the 2′ position: C.sub.1 to C.sub.10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH.sub.3, OCN, Cl, Br, CN, CF.sub.3, SOCH.sub.3, SO.sub.2CH.sub.3, ONO.sub.2, NO.sub.2, N.sub.3, NH.sub.2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an dsRNA, or a group for improving the pharmacodynamic properties of an dsRNA, and other substituents having similar properties. A preferred modification includes 2′-methoxyethoxy (2′-O—CH.sub.2CH.sub.2OCH.sub.3, also known as 2′-O-(2)methoxyethyl) or 2′-MOE) (Martin et al., Helv. Chim. Acta, 1995, 78, 486-504) i.e., an alkozy-alkoxy group. A further preferred modification includes 2′-dimethylaminooxyethoxy, i.e., a O(CH.sub.2).sub.2ON(CH.sub.3).sub.2 group, also known as 2′-DNAOE, as described in examples hereinbelow, and 2′-dimethylaminothoxyethoxy (also known in the art as 2′-O-dimethylaminoethoxyethyl or 2′-DMAEOE), i.e., 2′-O—CH.sub.2-O—CH.sub.2-N(CH.sub.2).sub.2, also described in examples hereinbelow.


Other preferred modifications include 2′-methoxy (2′-OCH.sub.3), 2′-aminopropoxy (2′-OCH.sub.2CH.sub2NH.sub.2) and 2′-fluoro (2′-F). Similar modifications may also be made at other positions on the dsRNA, particularly the 3′ position of the sugar on the 3′ terminal nulceotide or in 2′-5′ linked dsRNAs and the 5′ position of 5′ terminal nucleotide. DsRNAs may also have sugar mimetics such as cyclobutul moieties in place of the pentofuranosyl sugar. Representative U.S. patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; and 5,700,920, certain of which are commonly owned with the instant application, and each of which is herein incorporated by reference in its entirety.


DsRNAs may also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions. As used herein, “unmodified” or “natural” nucleobases include the puring bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouacil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl anal other 8-substituted adenines and guanines, 5-halo, particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and sytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-daazaadenine and 3-deazaguanine and 3-deazaadenin. Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. L, ed. John Wiley & Sons. 1990, these disclosed by Englisch et al., Angewandti Chemie, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y S., Chapter 15, DsRNA Research and Applications, pages 289-302, Crooke, S. T. and Lebleu, B., Ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynyleytosine, 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2.degree. C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., Eds., DsRNA Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are presently preferred base substitutions, even more particularly when combined with 2′-O-methoxyethyl sugar modifications.


Representative U.S. patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Pat No. 3,687,808, as well as U.S. Pat. Nos. 4,845,205; 5,130,30; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; and 5,681,941, each of which is herein incorporated by reference, and U.S. Pat. No. 5,750,692, also herein incorporated by reference.


Another modification of the dsRNAs of the invention involves chemically linking to the dsRNA one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the dsRNA. Such moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acid. Sci. USA, 199, 86, 6553-6556), cholic acid (Mancharan et al., Biorg. Med. Chem. Let., 1994 4 1053-1060), a thioether, e.g., beryl-S-triylthiol (Manoharan et al., Ann, N.Y. Acad. Sci., 1992, 660, 306-309; Manoharan et al., Biorg. Med. Chem. Let., 1993, 3, 2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20, 533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J, 1991, 10, 1111-1118; Kabanov et al., FEBS Lett., 1990, 259, 327-330; Svinarchuk et al., Biochimic, 1993, 75, 49-54), a phospholipid, e.g., di-hexadecyl-rac-hlycerol or triethyl-ammonium 1,2-di-O-hexadecyl-rac-glycero-3-Hphosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654; Shen et al., Nucl. Acids Res., 1990, 18, 3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237), or an octadecylamine or hexylamino-carbonyloxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923-937).


Representative U.S. patents that teach the preparation of such dsRNA conjugates include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538, 5,578,717, 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 5,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,345,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241; 5,391,723; 5,416,203; 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941, each of which is herein incorporated by reference.


It is not necessary for all positions in a given compound to be uniformly modified, and in fact more than one of the aforementioned modifications may be incorporated in a single compound or even at a single nucleoside within an dsRNA. The present invention also includes dsRNA compounds which are chimeric compounds. “Chimeric” dsRNA compounds or “chimeras,” in the context of this invention, are dsRNA compounds, particularly dsRNAs, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an dsRNA compound. These dsRNAs typically contain at least one region wherein the dsRNA is modified so as to confer upon the dsRNA increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid. An additional region of the dsRNA may serve as a substrate for anzymes capable of cleaving RNA-DNA or RNA:RNA hybrids. By way of example, RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNAduplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of dsRNA inhibition of gene expression. Consequently, comparable results can often be obtained with shorter dsRNAs when chimeric dsRNAs are used, compared to phosphorothioate deoxydsRNAs hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.


In certain instances, the dsRNA may be modified by a non-ligand group. A number of non-ligand molecules have been conjugated to dsRNAs in order to enhance the activity, cellular distribution or cellular uptake of the dsRNA, and procedures for performing such conjugations are available in the scientific literature. Such non-ligand moieties have included lipid moieties, such as cholesterol (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86:6553), cholic acid (Manoharan et al., Bioorg. Med. Chem. Lett., 1994, 4:1053), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad, Sci., 1992, 660-306; Manoharan et al., Bioorg. Med. Chem. Let. 1993, 3:2765), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20:533), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J., 1991, 10:111; Kabanov et al., FEBS Lett., 1990, 259:327; Svinarchuk et al., Biochimie, 1993, 75:49), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-O-hexadecyl-rac-hlycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36:3651; Shen et al., Nucl. Acids Res., 1990, 18:3777), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651), a palmityl moiety (Mishra et al., Biochim, Biophys. Acta, 1995, 1264:229), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacel. Exp. Ther., 1996, 277:923). Representative United States patents that teach the preparation of such dsRNA conjugates have been listed above. Typical conjugation protocols involve the synthesis of dsRNAs 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 dsRNA still bound to the solid support or following cleavage of the dsRNA in solution phase. Purification of the dsRNA conjugate by HPLC typically affords the pure conjugate.


Vector Encoded RNAi Agents


The dsRNA of the invention can also be expressed from recombinant viral vectors intracellularly in vivo. The recombinant viral vectors of the invention comprise sequences encoding the dsRNA of the invention and any suitable promoter for expressing the dsRNA sequences. Suitable promoters include, for example, the U6 or H1 RNA pol III promoter sequences and the cytomegalovirus promoter. Selection of other suitable promoters is within the skill in the art. The recombinant viral vectors of the invention can also comprise inducible or regulatable promoters for expression of the dsRNA in a particular tissue or in a particular intracellular environment. The use of recombinant viral vectors to deliver dsRNA of the invention to cells in vivo is discussed in more detail below.


dsRNA of the invention can be expressed from a recombinent viral vector either as two separate, complementary RNA molecules, or as a single RNA molecule with two complementary regions.


Any viral vector capable of accepting the coding sequences for the dsRNA molecule(s) so be expressed can be used, for example vectors derived from adenovirus (AV); adeno-associated virus (AAV); retroviruses (e.g, lentiviruses (LV), Rhabdoviruses, murine leukemia virus); harpes virus, and the like. The tropism of viral vectors can be modified by pseudotyping the vectors with envelope proteins or other surface antigens from other viruses, or by substituting different viral capsid proteins, as appropriate.


For example, lentiviral vectors of the invention can be pseudotyped with surface proteins from vesicular stomatitis virus (VSV), rabies, Ebola, Mokela, and the like. AAV vectors of the invention can be made to target different cells by engineering the vectors to express different capsid protein serotypes. For example, an AAV vector expressing a serotype 2 capsid on a serotype 2 genome is called AAV 2/2. This serotype 2 capsid gene in the AAV 2/2 vector can be replaced by a serotype 5 capsid gene to produce an AAV 2/5 vector. Techniques for constructing AAV vectors which express different capsid protein serotypes are within the skill in the art; see, e.g., Rabinowitz J E et al. (2002), J Virol 76:791-801, the entire disclosure of which is herein incorporated by reference.


Selection of recombinant viral vectors suitable for use in the invention, methods for inserting nucleic acid sequences for expressing the dsRNA into the vector, and methods of delivering the viral vector to the cells of interest are within the skill in the art. See, for example, Dornburg R (1995), Gene Therap. 2: 301-310, Eglitis M A (1988), Biotechniques 6: 608-614; Miller A D (1990), Hum Gene Therap. 1: 5-14; Anderson W F (1998), Nature 392: 25-30; and Rubinson D A et al., Nat. Genet. 33: 401-406, the entire disclosures of which are herein incorporated by reference.


Preferred viral vectors are those derived from AV and AAV. In a particularly preferred embodiment, the dsRNA of the invention is expressed as two separate, complementary single-stranded RNA molecules from a recombinant AAV vector comprising, for example, either the U6 or H1 RNA promoters, or the cytomegalovirus (CMV) promoter.


A suitable AV vector for expressing the dsRNA of the invention, a method for constructing the recombinant AV vector, and a method for delivering the vector into target cells, are described in Xia H et al. (2002), Nat. Biotech. 20: 1006-1010.


Suitable AAV vectors for expressing the dsRNA of the invention, methods for constructing the recombinant AV vector, and methods for delivering the vectors into target cells are described in Samulski R et al. (1987), J. Virol. 61: 3096-3101; Fisher K. J et al. (1996), J. Virol, 70: 520-532; Samulski R et al. (1989), J. Virol. 63: 3822-3826; U.S. Pat. No. 5,252,479; U.S. Pat. No. 5,139,941; International Patent Application No. WO 94/13788; and International Patent Application No. WO 93/24641, the entire disclosures of which are herein incorporated by reference.


III. Pharmaceutical Compositions Comprising dsRNA


In one embodiment, the invention provides pharmaceutical compositions comprising a dsRNA, as described herein, and a pharmaceutically acceptable carrier. The pharmaceutical composition comprising the dsRNA is useful for treating a disease or disorder associated with the expression or activity of the Eg5 gene, such as pathological processes mediated by Eg5 expression. Such pharmaceutical composition are formulated based on the mode of delivery. One example is compositions that are formulated for systemic administration via parenteral delivery.


In another embodiment, such compositions will further comprise a second dsRNA that inhibits VEGF expression. dsRNA directed to VEGF are described in the Examples and in co-pending U.S. Ser. Nos. 11/078,073 and 11/340,080.


The pharmaceutical compositions of the invention are administered to dosages sufficient to inhibit expression of the Eg5 gene (and VEGF expression when a second dsRNA is included). In general, a suitable dose of dsRNA will be in the range of 0.01 to 5.0 milligrams per kilogram body weight of the recipient per day, generally in the range of 1 microgram to 1 mg per kilogram body weight per day. The pharmaceutical composition may be administered once daily or the dsRNA may be administered as two, three, or more sub-doses at appropriate intervals throughout the day or even using continuous infusion or delivery through a controlled release formulation. In that case, the dsRNA contained in each sub-dose must be correspondingly smaller in order to achieve the total daily dosage. The dosage unit can also be compounded for delivery over several days, e.g., using a conventional sustained release formulation which provides sustained release of the dsRNA over a several day period. Sustained release formulations are well known in the art and are particularly useful for delivery of agents at a particular site, such as could be used with the agents of the present invention. In this embodiment, the dosage unit contains a corresponding multiple of the daily dose.


The skilled artisan will appreciate that certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or a series of treatments. Estimates of effective dosages and in vivo half-lives for the individual dsRNAs encompassed by the invention can be made using conventional methodologies or on the basis of in vivo testing using an appropriate animal model, as described elsewhere herein.


Advances in mouse genetics have generated a number of mouse models for the study of various human diseases, such as pathological processes mediated by Eg5 expression. Such models are used for in vivo testing of dsRNA, as well as for determining a therapeutically effective dose.


The present invention also includes pharmaceutical compositions and formulations which include the dsRNA compounds of the invention. The pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical, pulmonary, e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration.


Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves and the like may also be useful. Preferred topical formulations include those in which the dsRNAs of the invention are in admixture with a topical delivery agent such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents and sufactants. Preferred lipids and liposomes include neutral (e.g. dioleoylphosphatidyl DOPE ethanolamine, dimyristoylphosphatidyl choline DMPC, distearolyphosphatidyl choline) negative (e.g. dimyristoylphosphatidyl glycerol DMPG) and cationic (e.g. dioleoyltetramethylaminopropyl DOTAP and dioleoylphosphatidyl ethanolamine DOTMA). DsRNAs of the invention may be encapsulated within liposomes or may form complexes thereto, in particular to cationic liposomes. Alternatively, dsRNAs may be complexed to lipids, in particular to cationic lipids. Preferred fatty acids and esters include but are not limited arachidonic acid, oleic acid, eicosanoic acid, lauric acid, caprylic acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dieaprate, tricaprate, monoolein, dilaurin, glyceryl 1-monocaprate, 1-dodecylazacycloheptan-2-one, an acylcamitine, an acylcholine, or a C1-10 alkyl ester (e.g. isopropylmyristate IPM), monoglyceride, diglyceride or pharmaceutically acceptable salt thereof. Topical formulations are described in detail in U.S. patent application Ser. No. 09/315,298 filed on May 20, 1999 which is incorporated herein by reference in its entirety.


Compositions and formulations for oral administration include powders or granules, microparticulates, nanoparticulates, suspensions or solutions or solutions in water or non-aqueous media, capsules, gel capsules, sachets, tablets or minitablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable. Preferred oral formulations are those in which dsRNAs of the invention are administered in conjunction with one or more penetration enhancers surfactants and chelators. Preferred surfactants include fatty acids and/or esters or salts thereof, bile acids and/or salts thereof. Preferred bile acids/salts include chenodeoxycholic acid (CDCA) and ursodeoxychenodeoxycholic acid (UDCA), cholic acid, dehydrocholic acid, deoxycholic acid, glucholic acid, glycholic acid, glycodeoxycholic acid, taurocholic acid, taurodeoxycholic acid, sodium tauro-24,25-digydro-fusidate and sodium glycodihydrofusidate. Preferred fatty acids include arachidonic acid, undecanoic acid, oleic acid, lauric acid, caprylic acid, capric acid, myristic acid palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein, dilaurin, glycerly 1-monocaprate, 1-dodecylazacycloheptan-2-one, an acylcarnitine, an acylcholine, or a monoglyceride, a diglyceride or a pharmaceutically acceptable salt thereof (e.g. sodium). Also preferred are combinations of penetration enhancers, for example, fatty acids/salts in combination with bile acids/salts. A particularly preferred combination is the sodium salt of lauric acid, capric acid and UDCA. Further penetration enhancers include polyoxyethylene-9-lauryl ether, polyoxyethylene-20-cetyl ether. DsRNAs of the invention may be delivered orally, in granular form including sprayed dried particles, or complexed to form micro or nanoparticles. DsRNA complexing agents include poly-amino acids; polyimines; polyacrylates; polyalkylacrylates, polyoxethanes, polyalkyleyanoacrylates; cationized gelatins, albumins, starches, acrylates, polyethyleneglycols (PEG) and starches; polyalkylcyanoacrylates; DEAE-derivatized polimines, pollulans, celluloses and starches. Particularly preferred complexing agents include chitosan, N-trimethylchitosan, poly-L-lysine, polyhistidine, polyomithine, polyspermines, protamine, polyvinylpyridine, polythiodiethylaminomethylethylene P(TDAE), polyaminostyrene (e.g. p-amino), poly(methyleyanoacrylate), poly(ethylcyanoacrylate), poly(butylcayanoacrylate), poly(isobutylcyanoacrylate), poly(isohexyleynaoacrylate), DEAE-methacrylate, DEAE-hexylacrylate, DEAE-acrylamide, DEAE-albumin and DEAE-dextran, polymethylacrylate, polyhexylacrylate, poly(D,L-lactic acid), poly(DL-lactic-co-glycolic acid (PLGA), alginate, and polyethyleneglycol (PEG). Oral formulations for dsRNAs and their preparation are described in detail in U.S. application Ser. No. 08/886,829 (filed Jul. 1, 1997), Ser. No. 09/108,673 (filed Jul. 1, 1998), Ser No. 09/256,515 (filed Feb. 23, 1999), Ser. No. 09/082,624 (filed May 21, 1998) and Ser. No. 09/315,298 (filed May 20, 1999), each of which is incorporated herein by reference in their entirety.


Compositions and formulations for parenteral, intrathecal, intraventricular or intrahepatic administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients.


Pharmaceutical compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be generated from a variety of components that include, but are not limited to, preformed liquids, self-emulsifying solids and self-emulsifying semisolids. Particularly preferred are formulations that target the liver when treating hepatic disorders such as hepatic carcinoma.


The pharmaceutical formulations of the present invention, which may conventeitly be presented in unit dosage form, may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and the, if necessary, shaping the product.


The compositions of the present invention may be formulated into any of many possible dosage forms such as, but not limited to, tablets, capsules, gel capsules, liquid syrups, soft gels, suppositories, and enemas. The compositions of the present invention may also be formulated as suspensions in aqueous, non-aqueous or mixed media. Aqueous suspensions may further contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The suspension may also contain stabilizers.


Emulsions


The compositions of the present invention may be prepared and formulated as emulsions. Emulsions are typically heterogenous systems of one liquid dispersed in another in the form of droplets usually exceeding 0.1.mu.m in diameter (Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Bander (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volumn 1, p. 199; Rosoff, in Pharmaceutical Dosage Forms, Leiberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, INc., New York, N.Y., Volume 1, p. 245; Block in Pharmaceutical Dosage Forms, Leiberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., Volume 2, p. 335; Higuchi et al., in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 1985, p. 301). Emulsions are often biphasic systems comprising two immiscible liquid phases intimately mixed and dispersed with each other. In general emulsions may be of either the water-in-oil (w/o) or the oil-in-water (o/w) variety. When an aqueous phase is finely divided into and dispersed as minute droplets into a bulk oily phase, the resulting composition is called a water-in-oil (w/o) emulsion. Alternatively, when an oily phase is finely divided into and dispersed as minute droplets into a bulk aqueous phase, the resulting composition is called an oil-in-water (o/w) emulsion. Emulsions may contain additional components in addition to the dispersed phases, and the active drug which may be present as a solution in either the aqueous phase, oily phase or itself as a separate phase. Pharmaceutical excipients such as emulsifiers, stabilizers, dyes, and anti-oxidants may also be present in emulsions as needed. Pharmaceutical emulsions may also be multiple emulsions that are comprised of more than two phases such as, for example, in the case of oil-in-water-in-oil (o/w/o) and water-in-oil-in-water (w/o/w) emulsions. Such complex formulations often provide certain advantages that simple binary emulsions do not. Multiple emulsions in which individual oil droplets of an o/w emulsion enclose small water droplets constitute a w/o/w emulsion. Likewise a system of oil droplets enclosed in globules of water stabilized in an oily continuous phase provides an o/w/o emulsion.


Emulsions are characterized by little or no thermodynamic stability. Often, the dispersed or discontinuous phase of the emulsion is well dispersed into the external or continuous phase and maintained in this form through the means of emulsifiers or the viscosity of the formulation. Either of the phases of the emulsion may be a semisolid or a solid, as is the case of emulsion-style ointment bases and creams. Other means of stabilizing emulsions entail the use of emulsifiers that may be incorporated into either phase of the emulsion. Emulsifiers may broadly be classified into four categories: synthetic surfactants, naturally occurring emulsifiers, absorption bases, and finely dispersed solids (Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marchel Dekker, Inc., New York, N.Y., volume 1, p. 199).


Synthetic surfactants, also known as surface active agents, have found wide applicability in the formulation of emulsions and have been reviewed in the literature (Rieger, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 285; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), Marcel Dekker, Inc., New York, N.Y., 1988, volumn 1, p. 199). Surfactants are typically amphiphilic and comprise a hydrophilic and a hydrophobic portion. The ratio of the hydrophilic to the hydrophobic nature of the surfactant has been termed the hydrophile/lipophile balance (HLB) and is a valuable tool in categorizing and selecting surfactants in the preparation of formulations. Surfactants may be classified into different classes based on the nature of the hydrophilic group: nonionic, anionic, cationic and amphoteric (Rieger, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 285).


Naturally occurring emulsifiers used in emulsion formulations include lanolin, beeswax, phosphatides, lecithin and acacia. Absorption bases possess hydrophilic properties such that they can soak up water to form w/o emulsions yet retain their sermisolid consistencies, such as anhydrous lanolin and hydrophilic petrolatum. Finely divided solids have also been used as good emulsifiers especially in combination with surfactants and in viscous preparations. These include polar inorganic solids, such as heavy metal hydroxides, nonswelling clays such as bentonite, attapulgite, hectorite, kaolin montmorillonite, colloidal aluminum silicate and colloidal magnesium aluminum silicate, pigments and nonpolar solids such as carbon or glyceryl tristearate.


A large variety of non-emulsifying materials are also included in eulsion formulations and contribute to the properties of emulsions. These include fats, oils, waxes, fatty acids, fatty alcohols, fatty esters, humectants, hydrophilic colloids, preservatives and antioxidants (Block, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 335; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Bander (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199).


Hydrophilic colloids or hydrocolloids include naturally occurring gums and synthetic polymers such as polysaccharides (for example, acacia, agar, alginic acid, carrageenan, guar gum, karaya gum, and tragacanth), cellulose derivatives (for example, carboxymethylcellulose and carboxypropylcellulose), and synthetic polymers (for example, carbomers, cellulose ethers, and carboxyvinyl polymers). These disperse or swell in water to form colloidal solutions that stabilize emulsions by forming strong interfacial films around the dispersed-phase droplets and by increasing the viscosity of the ixternal phase.


Since emulsions often contain a number of ingredients such as carbohydrates, proteins, sterols and phosphatides that may readily support the growth of microbes, these formulations often incorporate preservatives. Commonly used preservatives included in emulsion formulations include methyl paraben, propyl paraben, quatemary ammonium salts, benzalkonium chloride, esters of p-hydroxybenzoic acid, and boric acid. Antioxidants are also commonly added to emulsion formulations to prevent detreioration of the formulation. Antioxidants used may be free radical scavengers such as tocopherols, alkyl gallates, butylated hydroxyanisole, butylated hydroxytoluene, or reducing agents such as ascorbic acid and sodium metabisulite, and antioxidant synergists such as citric acid, tartaric acid, and lecithin.


The application of emulsion formulations via dermatological, oral and parenteral routes and methods for their manufacture have been reviewed in the literature (Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199). Emulsion formulations for oral delivery have been very widely used because of ease of formulation, as well as efficacy from an absorption and bioavailability standpoint (Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199). Mineral-oil base laxatives, oil-soluble vitamins and high fat nutritive preparations are among the materials that have commonly been administered orally as o/w emulsions.


In one embodiment of the present invention, the compositions of dsRNAs and nucleic acids are formulated as microemulsions. A microemulsion may be defined as a system of water, oil and amphiphile which is a single optically isotropic and thermodynamically stable liquid solution (Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245). Typically microemulsions are systems that are prepared by first dispersing an oil in an aqueous surfactant solution and then adding a sufficient amount of a fourth component, generally an intermediate chain-length alcohol to form a transparent system. Therefore, microemulsions have also been described as thermodynamically stable, isotropically clear dispersions of two immiscible liquids that are stabilized by interfacial films of surface-active moleculses (Leung and Shah, in: Controlled Release of Drugs: Polymers and Aggregate Systems, Rosoff, M., Ed., 1989, VCH Publishers, New York, pages 185-215). Microemulsions commonly are prepared via a combination of three to five components that include oil, water, surfactant, cosurfactant and electrolyte. Whether the microemulsion is of the water-in-oil (w/o) or an oil-in-water (o/w) type is dependent on the properties of the oil and surfactant used and on the structure and geometric packing of the polar heads and hydrocarbon tails of the surfactant molecules (Schott, in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 1985, p. 271).


The phenomenological approach utilizing phase diagrams has been extensively studied and has yielded a comprehensive knowledge, to one skilled in the art, of how to formulate microemulsions (Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245; Block, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 335). Compared to conventional emulsions, microemulsions offer the advantage of solubilizing water-insoluble drugs in a formulation of thermodynamically stable droplets that are formed spontaneously.


Surfactants used in the preparation of microemulsions include, but are not limited to, ionic surfactants, non-ionic surfactants, Brij 96, polyoxyehtylene oleyl ethers, plyglycerol fatty acid esters, tetraglycerol monolaurate (ML310), tetraglycerol monooleate (MO310), hexaglycerol monooleate (PO310), hexaglycerol pentaoleate (PO500), decaglycerol monocaprate (MCA750), decaglycerol monooleate (MO750), decaglycerol sequioleate (SO750), decaglycerol decaoleate (DAO750), alone or in combination with cosurfactants. The sosurfactant, usually a short-chain alcohol such as ethanol, 1-propanol, and 1-butanol, serves to increase the interfacial fluidity by penetrating into the surfactant film and consequently creating a disordered film because of the void space generated among surfactant molecules. Microemulsions may, however, be prepared without the use of cosurfactants and alcohol-free self-emulsifying microemulsion systems are known in the art. The aqueous phase may typically be, but is not limited to, water, an aqueous solution of the drug, glycerol, PEG300, PEG400, polyglycerols, propylene glycols, and derivatives of ethylene glycol. The oil phase may include, but is not limited to, materials such as Captex 300, Captex 355, Capmul MCM, fatty acid esters, medium chain (C8-C12) mono, di, and tri-glycerides, polyoxyethylated glyceryl fatty esters, fatty alcohols, polyglycolized glycerides, saturated polyglycolized C8-C10 glycerides, vegetable oils and silicone oil.


Microemulsions are particularly of interest from the standpoint of drug solubilization and the enhanced absorption of drugs. Lipis based microemulsions (both o/w and w/o) have been proposed to enhance the oral bioavailibility of drugs, including peptides (Constrantinides et al., Pharmaceutical Research, 1994, 11, 1385-1390; Ritschel, Meth. Find. Exp. Clin. Pharmacol., 1993, 13, 205). Microemulsions afford advantages of improved drug solubilization, protection of drug from enzymatic hydrolysis, possible enhancement of drug absorption due to surfactant-induced alterations in membrane fluidity and permeability, ease of preparation, ease of oral administration over solic dosage forms, improved clinical potency, and decreased toxicity (Constantinides et al.,Pharmaceutical Research, 1994, 11, 1385; Ho et al., J. Pharm. Sci., 1996, 85, 138-143). Often microemulsions may form spontaneously when their components are brought together at ambient temperature. This may be particularly advantageous when formulating thermolabile drugs, piptides or dsRNAs. Microemulsions have also been effective in the transdermal delivery of active components in both cosmetic and pharmaceutical applications. It is expected that the microemulsion compositions and formulations of the present invention will facilitate the increased systemic absorption of dsRNAs and nucleic acids from the gastrointestinal tract, as well as improve the local cellular uptake of dsRNAs and nucleic acids.


Microemulsions of the present invention may also contain additional components and additives such as sorbitan monostearate (Grill 3), Labrasol, and penetration enhancers to improve the properties of the formulation and to enhance the absorption of the dsRNAs and nucleic acids of the present invention. Penetration enhancers used in the microemulsions of the present invention may be classified as belonging to one of five broad categories—surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92). Each of these classes has been discussed above.


Liposomes


There are many organized surfactant structures besides microemulsions that have been studied and used for the formulation of drugs. These include monolayers, micelles, bilayers and vesicles. Vesicles, such as liposomes, have attracted great interest because of their specificity and the duration of action they offer from the standpoint of drug delivery. As used in the present invention, the term “liposome” means a vesicle composed of amphiphilic lipids arranged in a spherical bilayer or bilayers.


Liposomes are unilamellar or multilamellar vesicles which have a membrane formed from a lipophilic material and an aqueous interior. The aqueous portion contains the composition to be delivered. Cationic liposomes possess the advantage of being able to fuse to the cell wall. Non-cationic liposomes, although not able to fuse as efficiently with the cell wall, are taken up by macrophages in vivo.


In order to cross intact mammalian skin, lipid vesicles must pass through a series of fine pores, each with a diameter less than 50 nm, under the influence of a suitable transdermal gradient. Therefore, it is desirable to use a liposome which is highly deformable and able to pass through such fine pores.


Further advantages of liposomes include; liposomes obtained from natural phospholipids are biocompatible and biodegradable; liposomes can incorporate a wide range of water and lipid soluble drugs; liposomes can protect encapsulated drugs in their internal compartments from metabolism and degradation (Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245). Important considerations in the preparation of liposome formulations are the lipid surface charge, vesicle size and the aqueous volume of the liposomes.


Liposomes are useful for the transfer and delivery of active ingredients to the site of action. Because the liposomal membrane is structurally similar to biological membranes, when liposomes are applied to a tissue, the liposomes start to merge with the cellular membranes and as the merging of the liposome and cell progresses, the liposomal contents are emptied into the cell where the active agent may act.


Lipsomal formulations have been the focus of extensive investigation as the mode of delivery for many drugs. There is growing evidence that for topical administration, liposomes present several advantages over other formulations. Such advantages include reduced side-effects related to high systemic absorption of the administered drug, increased accumulation of the administered drug at the desired target, and the ability to administer a wide variety of drugs, both hydrophilic and hydrophobic, into the skin.


Several reports have detailed the ability of liposomes to deliver agents including high-molecular weight DNA into the skin. Compounds including analgesics, antibodies, hormones and high-molecular weight DNAs have been administered to the skin. The majority of applications resulted in the targeting of the upper epidermis.


Liposomes fall into two broad classes. Cationic liposomes are positively charged liposomes which interact with the negatively charged DNA molecules to form a stable complex. The positively charged DNA/liposome complex binds to the negatively charged cell surface and is internalized in an endosome. Due to the acidic pH within the endosome, the liposomes are ruptured, releasing their contents into the cell cytoplasm (Wang et al., Biochem. Biophys. Res. Commun., 1987, 147, 980-985).


Liposomes which are pH-sensitive or negatively-charged , entrap DNA rather than complex with it. Since both the DNA and the lipid are similarly charged, repulsion rather than complex formation occurs. Nevertheless, some DNA is entrapped within the aqueous interior of these liposomes. pH-sensitive liposomes have been used to deliver DNA encoding the thymidine kinase gene to cell monolayers in culture. Expression of the exogenous gene was detected in the target cells (Zhou et al., Journal of Controlled Release, 1992, 19, 269-274).


One major type of liposomal composition includes phospholipids other than naturally-derived phosphatidylcholine. Neutral liposome compositions, for example, can be formed from dimyristoyl phosphatidylcholine (DMPC) or dipalmitoyl phosphatidylcholine (DPPC). Anionic liposome compositions generally are formed from dimyristoyl phosphatidylglycerol, while anionic fusogenic liposomes are formed primarily from dioleoyl phosphatidylethanolamine (DOPE). Another type of liposomal composition is formed from phosphatidylcholine (PC) such as, for example, soybean PC, and egg PC. Another type of formed from mixtures of phospholipid and/or phosphatidylcholine and/or cholesterol.


Several studies have assessed the topical delivery of liposomal drug formulations to the skin. Application of liposomes containing interferon to guinea pig skin resulted in a reduction of skin herpes sores while delivery of interferon via other means (e.g. as a solution or as an emulsion) were ineffective (Weiner et al., Journal of Drug Targeting, 1992, 2, 405-410). Further, an additional study tested the efficacy of interferon administered as part of a liposomal formulation to the administration of interferon using an aqueous system, and concluded that the liposomal formulation was superior to aqueous administration (du Plessis et al., Antiviral Research, 1992, 18, 259-265).


Non-ionic liposomal systems have also been examined to determine their utility in the delivery of drugs to the skin, in particular systems comprising non-ionic surfactant and cholesterol. Non-ionic liposomal formulations comprising Novasome.TM.I (glyceryl dilaurate/cholesterol/po-lyoxyethylene-10-stearyl ether) and Novasome.TM.II (glyceryl distearate/cholesterol/polyoxyethylene-10-stearyl ether) were used to deliver cyclosporin-A into the dermis of mouse skin. Results indicated that such non-ionic liposomal systems were effective in facilitating the deposition of cyclosporin-A into different layers of the skin (Hu et al. S.T.P.Pharma. Sci., 1994, 4, 6, 466).


Liposomes also include “sterically stabilized” liposomes, a term which, as used herein, refers to liposomes comprising one or more specialized lipids that, when incorporated into liposomes, result in enhanced circulation lifetimes relative to liposomes lacking such specialized lipids. Examples of sterically stabilized liposomes are those in which part of the vesicle-forming lipid portion of the liposome (A) comprises one or more glycolipids, such as monosialoganglioside G.sub.M1, or (B) is derivatized with one or more hydrophilic polymers, such as a polyethylene glycol (PEG) moiety. While not wishing to be bound by any particular theory, it is thought in the art that, at least for sterically stabilized liposomes containing gangliosides, sphingomyelin, or PEG-derivatized lipids, the enhanced circulation half-life of these sterically stabilized liposomes derives from a reduced uptake into cells of the reticuloendothelial system (RES) (Allen et al., FEBS Letters, 1987, 223, 42; Wu et al., Cancer Research, 1993, 53, 3765).


Various liposomes comprising one or more glycolipids are known in the art. Papagadjopoulos et al. (Ann. N.Y. Acad. Sci., 1987, 507, 64) reported the ability of monosialoganglioside G.sub.M1, galactocerebrodside sulfate and phosphatidylinositol to improve blood half-lives of liposomes. These findings were expounded upon by Gabizon et al. (Proc. Natl. Acad. Sci. U.S.A., 1988, 85, 6949), U.S. Pat. No. 4,837,028 and WO 88/04924, both to Allen et al., disclose liposomes comprising (1) sphingomyelin and (2) the ganglioside G.sub.M1 or a galactocerebroside sulfate ester. U.S. Pat. No. 5,543,152 (Webb et al.) discloses liposomes comprising sphingomyelin. Liposomes comprising 1,2-sn-dimyristoylphosphat-idylcholine are disclosed in WO 97/13499 (Lim et al).


Many liposomes comprising lipids derivatized with one or more hydrophilic polymers, and methods of preparation thereof, are known in the art. Sunamoto et al. (Bull. Chem. Soc. Jpn., 1980, 53, 2778) described liposomes comprising a nonionic detergent, 2C.sub.1215G, that contains a PEG moiety. Illum et al. (FEBS Lett., 1984, 167, 79) noted that hydrophilic couting of polystyrene particles with polymeric glycols results in significantly enhanced blood half-lives. Synthetic phospholipids modified by the attachment of carboxylic groups of polyalkylene glycols (e.g., PEG) are described by Sears (U.S. Pat. Nos. 4,426,330 and 4,534,899). Klibanov et al. (FEBS Lett., 1990, 268, 235) described experiments demonstrating that liposomes comprising phosphatidylethanolamine (PE) derivatized with PEG or PEG stearate have significant increases in blood circulation half-lives. Blume et al. (Biochimica et Biophsica Acta, 1990, 1029, 91) extended such observations to other PEG-derivatized phospholipids, e.g., DSPE-PEG, formed from the combination of distearoylphosphatidylethanolamine (DSPE) and PEG.Liposomes having covalently bound PEG moieties on their external surface are described in European Patent No. EP 0 445 131 B1 and WO 90/04384 to Fisher. Liposome compositions containing 1-20 mole percent of PE derivatized with PEG, and methods of use thereof, are described by Woodle et al. (U.S. Pat. Nos. 5,013,556 and 5,356,633) and Martin et al. (U.S. Pat. No. 5,213,804 and European Patent No. EP 0 496 813 B1). Liposomes comprising a number of other lipid-polymer conjugates are disclosed in WO 91/05545 and U.S. Pat. No. 5,225,212 (both to Martin et al.) and in WO 94/20073 (Zalipsky et al.) Liposomes comprising PEG-modified ceramide lipids are described in WO 96/10391 (Choi et al). U.S. Pat. No. 5,540,935 (Miyazaki et al.) and U.S. Pat. No. 5,556,948 (Tagawa et al.) describe PEG-containing liposomes that can be further derivatized with functional moieties on their surfaces.


A limited number of liposomes comprising nucleic acids are known in the art. WO 96/40062 to Thierry et al. discloses methods for encapsulating high molecular weight nucleic acids in liposomes. U.S. Pat. No. 5,264,221 to Tagawa et al. discloses protein-bonded liposomes and asserts that the contents of such liposomes may include an dsRNA RNA. U.S. Pat. No. 5,665,710 to Rahman et al. describes certain methods of encapsulating oligodeoxynucleotides in liposomes. WO 97/04787 to Love et al. discloses liposomes comprising dsRNA dsRNAs targeted to the raf gene.


Transfersomes are yet another type of liposomes, and are highly deformable lipid aggregates which are attractive candidates for drug delivery vehicles. Transfersomes may be described as lipid droplets which are so highly deformable that they are easily able to penetrate through pores which are smaller than the droplet. Transfersomes are adaptable to the environment in which they are used, e.g. they are self-optimizing (adaptive to the shape of pores in the skin), self-repairing, frequently reach their targets without fragmenting, and often self-loading. To make transfersomes it is possible to add surface edge-activators, usually surfactants, to a standard liposomal composition. Transfersomes have been used to deliver serum albumin to the skin. The transfersome-mediated delivery of serum albumin has been shown to be as effective as subcutaneous injection of a solution containing serum albumin.


Surfactants find wide application in formulations such as emulsions (including microemulsions) and liposomes. The most common way of classifying and ranking the properties of the many different types of surfactants, both natural and synthetic, is by the use of the hydrophile/lipophile balance (HLB). The nature of the hydrophilic group (also known as the “head”) provides the most useful means for categorizing the different surfactants used in formulations (Rieger, in Pharmaceutical Dosage Forms, Marcel Dekker, Inc., New York, N.Y. 1988, p. 285).


If the surfactant molecule is not ionized, it is classified as a nonionic surfactant. Nonionic surfactants find wide application in pharmaceutical and cosmetic products and are usable over a wide range of pH values. In general their HLB values range from 2 to about 18 depending on their structure. Nonionic surfactants include nonionic esters such as ethylene glycol esters, propylene glycol esters, glyceryl esters, polyglyceryl esters, sorbitan esters, sucrose esters, and ethoxylated esters. Nonionic alkanolamides and ethers such as fatty alcohol ethoxylates, propoxylated alcohols, and ethoxylated/propoxylated block polymers are also included in this class. The polyoxyethylene surfactants are the most popular members of the nonionic surfactant class.


If the surfactant molecule carries a negative charge when it is disolved or dispersed in water, the surfactant is classified as anionic. Anionic surfactants include carboxylates such as soaps, acyl lactylates, acyl amides of amino acids, esters of sulfuric acid such as alkyl sufates and ethoxylated alkyl sulfates, sulfonates such as alkyl benzene sulfonates, acyl isethionates, acyl taurates and sulfosuccinates, and phosphates. The most important members of the anionic surfactant class are the alkyl sulfates and the soaps.


If the surfactant molecule carries a positve charge when it is dissolved or dispersed in water, the surfactant is classified as cationic. Cationic surfactants include quaternary ammonium salts and ethoxylated amines. The quaternary ammonium salts are the most used members of this class.


If the surfactant molecule has the ability to carry either a positive or negative charge, the surfactant is classified as amphoteric. Amphoteric surfactants include acrylic acid derivatives, substituted alkylamides, N-alkylbetaines and phosphatides.


The use of surfactants in drug products, formulations and in emulsions has been reviewed (Rieger, in Pharmaceutical Dosage Forms, Marcel Dekker, Inc., New York, N.Y., 1988, p. 285).


Penetration Enhancers


In one embodiment, the present invention employs various penetration enhancers to effect the efficient delivery of nucleic acids, particularly dsRNAs, to the skin of animals. Most drugs are present in solution in both ionized and nonionized forms. However, usually only lipid soluble or lipophilic drugs readily cross cell membranes. It has been discovered that even non-lipophilic drugs may cross cell membranes if the membrane to be crossed is treated with a penetration enhancer. In addition to aiding the diffusion of non-lipophilic drugs across cell membranes, penetration enhancers also enhance the permeability of lipophilic drugs.


Penetration enhancers may be classified as belonging to one of five broad categories, i.e., surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92). Each of the above mentioned classes of penetration enhancers are described below in greater detail.


Surfactants: In connection with the present invention, surfactants (or “surface-active agents”) are chemical entities which, when dissolved in an aqueous solution, reduce the surface tension of the solution or the interfacial tension between the aqueous solution and another liquid, with the result that absorption of dsRNAs through the mucosa is enhanced. In addition to bile salts and fatty acids, these penetration enhancers include, for example, sodium lauryl sulfate, polyoxyethylene-9-lauryl ether and polyoxyethylene-20-cetyl ether) (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92); and perfluouochemical emulsions, such as FC-43. Takahashi et al., J. Pharm. Pharmacol., 1988, 40, 252).


Fatty acids: Various fatty acids and their derivatives which act as penetration enhancers include, for example, oleic acid, lauric acid, capric acid (N-decanoic acid), myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein (1-monooleoyl-rac-glycerol), dilaurin, caprylic acid, arachidonic acid, glycerol 1-monocaprate, 1-dodecylazacycloheptan-2-one, acylearnitines, acylcholines, C.sub.1-10 alkyl esters thereof (e.g., methyl, isopropyl and t-butyl), and mono- and di-glycerides thereof (i.e., oleate, laurate, caprate, myristate, palmitate, stearate, linoleate, etc.) (Lee et al., Critical Reviews in Therapeutic Drug Carryier Systems, 1991, p. 92; Muranishi, Critical Reviews in Terapeutic Drug Carrier Systems, 1990, 7, 1-33; El Hariri et al., J. Pharm. Pharmacol., 1992, 44, 651-654).


Bile salts: The physiological role of bile includes the facilitation of dispersion and absorption of lipids and fat-soluble vitamins (Brunton, Chapter 38 in: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al. Eds., McGraw-Hill, New York, 1996, pp. 934-935). Various natural bile salts, and their synthetic derivatives, act as penetration enhancers. Thus the term “bile salts” includes any of the naturally occurring components of bile as well as any of their synthetic derivatives. The bile salts of the invention include, for example, cholic acid (or its pharmaceutically acceptable sodium salt, sodium cholate), dehydrocholic acid (sodium dehydrocholate), deoxycholic acid (sodium deoxycholate), glucholic acid (sodium glucholate), glycholic acid (sodium glycocholate), glycodeoxyeholic acid (sodium glycodeoxycholate), taurocholic acid (sodium taurocholate), taurodeoxycholic acid (sodium taurodeoxycholate), chanodeoxycholic acid (sodium chenodeoxycholate), ursodeoxycholic acid (UDCA), sodium tauro-24,25-dihydro-fusidate (STDHF), sodium glycodihydrofusidate and polyoxyethylene-9-lauryl ether (POE) (Lee et al., Critical Reviews in therapeutic Drug Carrier Systems, 1991, page 92; Swinyard, Chapter 39 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, Pa., 1990, pages 782-783; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 79, 579-583).


Chelating Agents: Chelating agents, as used in connection with the present invention, can be defined as compounds that remove metallic ions from solution by forming complexes therewith, with the result that absorption of dsRNAs through the mucosa is enhanced. With regards to their use as penetration enhancers in the present invention, chelating agents have the added advantage of also serving as DNase inhibitors, as most characterized DNA nucleases require a divalent metal ion for catalysis and are thus inhibited by chelating agents (Jarrett, J. Chromatogr., 1993, 618, 315-339). Chelating agents of the invention include but are not limited to disodium ethylenediaminetetraacetate (EDTA), citric acid, salicylates (e.g., sodium salicylate, 5-methoxysalicylate and homovanilate), N-acyl derivatives of collagen, laureth-9 and N-amino acyl derivatives of beta-diketones (enamines)(Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33; Buur et al., J. Control Rel., 1990, 14, 43-51).


Non-chelating non-surfactants: As used herein, non-chelating non-surfactant penetration enhancing compounds can be defined as compounds that demonstrate insignificant activity as chelating agents or as surfactants but that nonetheless enhance absorption of dsRNAs through the alimentary mucose (Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33). This class of penetration enhancers include, for example unsaturated cyclic ureas, 1-alkyl- and 1-alkenylazacycle-alkanone derivatives (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92); and non-steroidal anti-inflammatory agents such as diclofenac sodium, indomethacin and phenylbutazone (Yamashita et al., J. Pharm. Pharmacol., 1987, 39, 621-626).


Agents that enhance uptake of dsRNAs at the cellular level may also be added to the pharmaceutical and other compositions of the present invention. For example cationic lipids, such as lipodectin (Junichi et al, U.S. Pat. No. 5,705,188), cationic hlycerol derivatives, and polycationic molecules, such as polylysine (Lollo et al., PCT Application WO 97/30731), are also known to enhance the cellular uptake of dsRNAs.


Other agents may be utilized to enhance the penetration of the administered nucleic acids, including glycols such as ethylene glycol and propylene glycol, pyrrols such as 2-pyrrol, azones, and terpenes such as limonene and menthone.


Carriers


Certain compositions of the present invention also incorporate carrier compounds in the formulation. As used herein, “carrier compound” or “carrier” can refer to a nucleic acid, or analog thereof, which is inert (i.e., does not possess biological activity per se) but is recognized as a nucleic acid by in vivo processes that reduce the bioavailability of a nucleic acid having biological activity by, for example, degrading the biologically active nucleic acid or promoting its removal from circulation. The coadministration of a nucleic acid and a carrier compound, typically with an excess of the latter substance, can result in a substantial reduction of the amount of nucleic acid recovered in the liver, kidney or other extracirculatory reservoirs, presumably due to competition between the carrier compound and the nucleic acid for a common receptor. For example, the recovery of a partially phosphorothioate dsRNA in hepatic tissue can be reduced when it is coadministered with polyinosinic acid, dextran sulate, polyytidic acid or 4-acetamido-4′isothiocyano-stilbene-2,2′-disulfonic acid (Miyao et al., DsRNA Res. Dev., 1995, 5, 115-121; Takakura et al., DsRNA & Nucl. Acid Drug Dev., 1996, 6, 177-183.


Excipients


In contrast to a carrier compound, a “pharmaceutical carrier” or “excipient” is a pharmaceutically acceptable solvent, suspending agent or any other pharmacologically inert vehicle for delivering one or more nucleic acids to an animal. The excipient may be liquid or solic and is selected, with the planned manner of administration in mind, so as to provide for the desired bulk, consistency, etc., When combined with a nucleic acid and the other components of a given pharmaceutical composition. Typical pharmaceutical carriers include, but are not limited to, binding agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose, etc.); fillers (e.g., lactose and other sugars, microcrystalline cellulose, pectin, gelatin, calcium sulfate, ethyl cellulose, polyacrylates or calcium hydrogen phosphate, etc.); lubricants (e.g., magnesium stearate, tale, silica, colloidal silicon dioxide, stearic acid, metallic stearates, hydrogenated vegetable oils, corn starch, polyethylene glycols, sodium benzoate, sodium acetate, etc.); disintegrants (e.g., starch, sodium starch glycolate, etc.); and wetting agents (e.g., sodium lauryl sulphate, etc).


Pharmaceutically acceptable organic or inorganic excipient suitable for non-parenteral administration which do not deleteriously react with nucleic acids can also be used to formulate the compositions of the present invention. Suitable pharmaceutically acceptable carriers include, but are not limited to, water, salt solutions, alcohols, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose, polyvinylpyrrolidone and the like.


Formulations for toptical administration of nucleic acids may include sterile and non-sterile aqueous solutions, non-aqueous solutions in common solvents such as alcohols, or solutions of the nucleic acids in liquid or solid oil bases. The solutions may also contain buffers, diluents and other suitable additives. Pharmaceutically acceptable organic or inorganic excipients suitable for non-parenteral administration which do not deleteriously react with nucleic acids can be used.


Suitable pharmaceutically acceptable excipients include, but are not limited to, water, salt solutions, alcohol, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose, polyvinylpyrrolidone and the like.


Other Components


The compositions of the present invention may additionally contain other adjunct components conventionally found in pharmaceutical compositions, at their art-established usage levels. Thus, for example, the compositions may contain additional, compatible, pharmaceutically-active materials such as, for example, antipurities, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the compositions of the present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. However, such materials, when added, should not unduly interfere with the biological activities of the components of the compositions of the present invention. The formulations can be sterilized and, if desired, mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously interact with the nucleic acid(s) of the formulation.


Aqueous suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The suspension may also contain stabilizers.


Certain embodiments of the invention provide pharmaceutical compositions containing (a) one or more antisense compounds and (b) one or more other chemotherapeutic agents which function by a non-antisense mechanism. Examples of such chemotherapeutic agents include but are not limited to daunorubicin, daunomycin, dactinomycin, doxorubicin, epirubicin, idarubicin, esorubicin, bleomycin, mafosfamide, ifosfamide, cytosine arabinoside, bis-chloroethylnitrosurea, busulfan, mitomycin C, actinomycin D, mithramycin, prednisone, hydroxyprogesterone, testosterone, tamoxifen, decarbazine, procarbazine, hexamethylmelamine, pentamethylmelamine, mitoxantrone, amsacrine, chlorambucil, methylcyclohexylnitrosurea, nitrogen mustards, melphalan, cyclophosphamide, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-azacytidine, hydroxyurea, deoxycoformycin, 4-hydroxyperoxycyclophosphor-amide, 5-fluorouracil (5-FU), 5-fluorodeoxyuridine (5-FUdR), methotrexate (MTX), colchicine, taxol, vincristine, vinblastine, etoposide (VP-16), trimetrexate, irinotecan, topotecan, gemcitabine, teniposide, cisplatin and diethylstilbestrol (DES). See, generally, The Merck Manual of Diagnosis and Therapy, 15th Ed. 1987, pp. 1206-1228, Berkow et al., eds., Rahway, N.J. When used with the compounds of the invention, such chemotherapeutic agents may be used individually (e.g., 5-FU and oligonucleotide), sequentially (e.g., 5-FU and oligonucleotide for a period of time followed by MTX and oligonucleotide), or in combination with one or more other such chemotherapeutic agents (e.g., 5-FU, MTX and oligonucleotide, or 5-FU, radiotherapy and oligonucleotide). Anti-inflammatory drugs, including but not limited to nonsteroidal anti-inflammatory drugs and corticosteroids, and antiviral drugs, including but not limited to ribivirin, vidarabine, acyclovir and ganciclovir, may also be combined in compositions of the invention. See, generally, The Merck Manual of Diagnosis and Therapy, 15th Ed., Berkow et al., eds., 1987, Rahway, N.J., pages 2499-2506 and 46-49, respectively). Other non-antisense chemotherapeutic agents are also within the scope of this invention. Two or more combined compounds may be used together or sequentially.


Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the Ed50 (the dose therapeutically effective in 50% of the population). The dose ration between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds which exhibit high therapeutic indices are preferred.


The data obtained from cell culture assays and animal studies can be used in formulation a range of dosage for use in humans. The dosage of compositions of the invention lies generally within a range of circulating concentrations that include the Ed50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range of the compound or, when appropriate, of the polypeptide product of a target sequence (e.g., achieving a decreased concentration of the polypeptide) that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.


In addition to their administration individually or as a plurality, as discussed above, the dsRNAs of the invention can be administered in combination with other known agents effective in treatment of pathological processes mediated by Eg5 expression. In any event, the administering physician can adjust the amount and timing of dsRNA administration on the basis of results observed using standard measures of efficacy known in the art or described herein.


Methods for Treating Diseases Caused by Expression of the Eg5 Gene


The invention relates in particular to the use of a dsRNA or a pharmaceutical composition prepared thereform for the treatment of cancer, e.g., for inhibiting tumor growth and tumor metastasis. For example, the dsRNA or a pharmaceutical composition prepared therefrom may be used for the treatment of solid tumors, like breast cancer, lung cancer, head and neck cancer, brain cancer, abdominal cancer, colon cancer, colorectal cancer, esophagus cancer, gastrointestinal cancer, glioma, liver cancer, tongue cancer, neuroblastoma, osteosarcoma, ovarian cancer, pancreatic cancer, prostate cancer, retinoblastoma, Wilm's tumor, multiple myeloma and for the treatment of skin cancer, like melanoma, for the treatment of lymphormas and blood cancer. The invention further relates to the use of an dsRNA according to the invention or a pharmaceutical composition prepared therefrom for inhibiting eg5 expression and/or for inhibiting accumulation of ascites fluid and plural effusion in different types of cancer, e.g., breast cancer, lung cancer, head cancer, neck cancer, brain cancer, abdominal cancer, colon cancer, colorectal cancer, esophagus cancer, gastrointestinal cancer, glioma, liver cancer, tongue cancer, neuroblastoma, osteosarcoma, ovarian cancer, pancreatic cancer, prostate cancer, retinoblastoma, Wilm's tumor, multiple myeloma, skin cancer, melanoma, lymphomas and blood cancer. Owing to the inhibitory effect on eg5 expression, an dsRNA according to the invention or a pharmaceutical composition prepared thereform can enhance the quality of life.


The invention futhermore relates to the use of an dsRNA or a pharmaceutical composition thereof, e.g., for treating cancer or for preventing tumor metastasis, in combination with other pharmaceuticals and/or other therapeutic methods, e.g., with known pharmaceuticals and/or known therapeutic methods, such as, for example, those which are currently employed for treating cancer and/or for preventing tumor metastasis. Preference is given to a combination with radiation therapy and chemotherapeutic agents, such as cisplatin, cyclophosphamide, 5-fluoroacil, adriamycin, daunorabicin or tamoxifen. Other embodiments include the use of a second dsRNA used to inhibit the expression of VEGF.


The invention can also be practiced by including with a specific RNAi agent, in combination with another anti-cancer chemotherapeutic agent, such as any conventional chemotherapeutic: agent, or another dsRNA used to inhibit the expression of VEGF. The combination of a specific binding agent with such other agents can potentiate the chemotherapeutic protocol. Numerous chemotherapeutic protocols will present themselves in the mind of the skilled practitioner as being capable of incorporation into the method of the invention. Any chemotherapeutic agent can be used, including alkylating agents, antimetabolites, hormones and antagonists, radioisotopes, as well as natural products. For example, the compound of the invention can be administered with antibiotics such as doxorubicin and other anthracycline analogs, nitrogen mustards such as cyclophosphamide, pyrimidine analogs such as 5-fluorouracil, cisplatin, hydroxyurea, traxol and its natural and synthetic derivatives, and the like. As another example, in the case of mixed tumors, such as adenocarcinoma of the breast, where the tumors include gonadotropin-dependent and gonadotropin-independent cells, the compound can be administered in conjunction with leuprolide or goserelin (synthetic peptide analogs of LH-RH). Other antineoplastic protocols include the use of a tetracycline compound with another treatment modality, e.g.,surgery, radiation, etc., also referred to herein as “adjunct antineoplastic modalities.” Thus, the method of the invention can be employed with such conventional regimens with the benefit of reducing side effects and enhancing efficacy.


Methods for Inhibiting Expression of the Eg5 Gene


In yet another aspect, the invention provides a method for inhibiting the expression of the Eg5 gene in a mammal. The method comprises administering a composition of the invention to the mammal such that expression of the target Eg5 gene is silenced. Because of their high specificity, the dsRNAs of the invention specifically target RNAs (primary or processed) of the target Eg5 gene. Compositions and methods for inhibiting the expression of these Eg5 genes using dsRNAs can be performed as described elsewhere herein.


In one embodiment, the method comprises administering a composition comprising a dsRNA, wherein the dsRNA comprises a nucleotide sequence which is complementary to at least a part of an RNA transcript of the Eg5 gene of the mammal to be treated. When the organism to be treated is a mammal such as a human, the compositions may be administered by any means known in the art including, but not limited to oral or parenteral routes, including intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), nasal, rectal, and topical (including buccal and sublingual) administration. In preferred embodiments, the compositions are administered by intravenous infusion or injection.


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. All 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.


EXAMPLES

Gene Walking of the Eg5 Gene


Initial Screening Set


siRNA design was carried out to identify siRNAs targeting Eg5 (also known as KIF11, HSKP, KNSL1 and TRIP5). Human mRNA sequences to Eg5, RefSeq ID number:NM004523, was used.


siRNA duplexes cross-reactive to human and mouse Eg5 were designed. Twenty-four duplexes were synthesized for screening. (Table 1).


Expanded Screening Set


A second screening set was defined with 266 siRNAs targeting human EG5, as well as its rhesus monkey ortholog (Table 2). An expanded screening set was selected with 328 siRNA targeting human EG5, with no necessity to hit any EG5 mRNA of other species (Table 3).


The sequences for human and a partial rhesus EG5 mRNAs were downloaded from NCBI Nucleotide database and the human sequence was further on used as reference sequence (Human EG5:NM004523.2, 4908 bp, and Rhesus EG5: XM001087644.1, 878 bp (only 5′ part of human EG5).


For identification of further rhesus EG5 sequences a mega blast search with the human sequence was conducted at NCBI against rhesus reference genome. The downloaded rhesus sequence and the hit regions in the blast hit were assembled to a rhesus consensus sequence with ˜92% identity to human EG5 over the full-length.


All possible 19 mers were extracted from the human mRNA sequence, resulting in the pool of candidate target sites corresponding to 4890 (sense strand) sequences of human-reactive EG5 siRNAs.


Human-rhesus cross-reactivity as prerequisite for in silico selection of siRNAs for an initial screening set out of this candidate pool. To determine rhesus-reactive siRNAs, each candidate siRNA target site was searched for presence in the assembled rhesus sequence. Further, the predicted specificity of the siRNA as criterion for selection of out the pool of human-rhesus cross-reactive siRNAs, manifested by targeting human EG5 mRNA sequences, but not other human mRNAs.


The specificity of an siRNA can be expressed via its potential to target other genes, which are referred to as “off-target genes”.


For predicting the off-target potential of an siRNA, the following assumptions were made:

    • 1) off-target potential of a strand can be deduced from the number and distribution of mismatches to an off-target
    • 2) the most relevant off-target, that is the gene predicted to have the highest probability to be silenced due to tolerance of mismatches, determines the off-target potential of the strand
    • 3) positions 2 to 9 (counting 5′ to 3′) of a strand (seed region) may contribute more to off-target potential than rest of sequence (that is non-seed and cleavage site region)
    • 4) positions 10 and 11 (counting 5′ to 3′) of a strand (cleavage site region) may contribute more to off-target potential than non-seed region (that is positions 12 to 18, counting 5′ to 3′)
    • 5) position 1 and 19 of each strand are not relevant for off-target interactions
    • 6) off-target potential can be expressed by the off-target score of the most relevant off-target, calculated based on number and position of mismatches of the strand to the most homologous region in the off-target gene considering assumptions 3 to 5
    • 7) off-target potential of antisense and sense strand will be relevant, whereas potential abortion of sense strand activity by internal modifications introduced is likely


SiRNAs with low off-target potential were defined as preferable and assumed to be more specific.


In order to identify human EG5-specific siRNAs, all other human transcripts, which were all considered potential off-targets, were searched for potential target regions for human-rhesus cross-reactive 19 mer sense strand sequences as well as complementary antisense strands. For this, the fastA algorithm was used to determine the most homologues hit region in each sequence of the human RefSeq database, which we assume to represent the comprehensive human transcriptome.


To rank all potential off-targets according to assumptions 3 to 5, and by this identify the most relevant off-target gene and its off-target score, fastA output files were analyzed further by a perl script.


The script extracted the following off-target properties for each 19 mer input sequence and each off-target gene to calculate the off-target score:


Number of mismatches in non-seed region


Number of mismatches in seed region


Number of mismatches in cleavage site region


The off-target score was calculated by considering assumptions 3 to 5 as follows:







Off


-


target





score

=


number





of





seed





mismatches
*
10

+

number





of





cleavage





site





mismatches
*
1.2

+

number





of





non


-


seed





mismatches
*
1






The most relevant off-target gene for each 19 mer sequence was defined as the gene with the lowest off-target score. Accordingly, the lowest off-target score was defined as representative for the off-target potential of a strand.


For the screening set in Table 2, an off-target score of 3 or more for the antisense strand and 2 or more for the sense strand was chosen as prerequisite for selection of siRNAs, whereas all sequences containing 4 or more consecutive G's (poly-G sequences) were excluded. 266 human-rhesus cross-reactive sequences passing the specificity criterion, were selected based on this cut-off (see Table 2).


For definition of the expended screening set the cross-reactivity to rhesus was disgarded, re-calculated the predicted specificity based on the newly available human RefSeq database and selected only those 328 non-poly-G siRNAs with off-target score of 2,2 or more for the antisense and sense strand (see Table 3).


For the Tables: Key: A,G,C,U-ribonucleotides: T-deoxythymidine: u,c-2′-O-mehtyl nucleotides: s-phosphorothioate linkage


dsRNA Synthesis


Source of Reagents


Where the source of a reagent is not specifically given herein, such reagent may be obtained from any supplier of reagents for molecular biology at a quality/purity standard for application in molecular biology.


siRNA Synthesis


Single-stranded RNAs were produced by solid phase synthesis on a scale of 1 μmole using an Expedite 8909 synthesizer (Applied Biosystems, Applera Deutschland GmbH, Darmstadt, Germany) and controlled pore glass (CPG, 500 Å, Proligo Biochemie GmbH, Hamburg, Germany) as solid support. RNA and RNA containing 2′-O-methyl nucleotides were generated by solid phase synthesis employing the corresponding phosphoramidites and 2′-O-methyl phosphoramidites, respectively (Proligo Biochemic GmbH, Hamburg, Germany). These building blocks were incorporated at selected sites within the sequence of the oligoribonucleotide chain using standard nucleoside phosphoramidite chemistry such as descried in Current protocols in nucleic acid chemistry, Beaucage, S. L. et al. (Edrs.), John Wiley & Sons, Ind., New York, N.Y., USA. Phosphorothioate linkages were introduced by replacement of the iodine oxidizer solution with a solution of the Beaucage reagent (Chruachem Ltd, Glasgow, UK) in acetonitrile (1%). Further ancillary reagents were obtained from Mallinckrodt Baker (Griesheim, Germany).


Deprotection and purification of the crude oligoribonucleotides by anion exchange HPLC were carried out according to established procedures. Yields and concentrations were determined by UV absorption of a solution of the respective RNA at a wavelength of 260 nm using a spectral photometer (DU 640B, Beckman Coulter GmbH, Unterschleiβheim, Germany). Double stranded RNA was generated by mixing an equimolar solution of complementary strands in annealing buffer (20 mM sodium phosphate, pH 6.8; 100 mM sodium chloride), beated in a water bath at 85-90° C. for 3 minutes and cooled to room temperature over a period of 3-4 hours. The annealed RNA solution was stored at −20° C. until use.


For the synthesis of 3′-cholesterol-conjugated siRNAs (herein referred to as -Chol-3′), an appropriately modified solid support was used for RNA synthesis. The modified solid support was prepared as follows:


Diethyl-2-azabutane-1,4-dicarboxylate AA



embedded image


A 4.7 M aqueous solution of sodium hydroxide (50 mL) was added into a stirred, ice-cooled solution of ehtyl glycinate hydrochloride (32.19 g, 0.23 mole) in water (50 mL). Then, ethyl acrylate (23.1 g, 0.23 mole) was added and the mixture was stirred at room temperature until completion of the reaction was ascertained by TLC. After 19 h the solution was partitioned with dichloromethane (3×100 mL). The organic layer was dried with anhydrous sodium sulfate, filtered and evaporated. The residue was distilled to afford AA (28.8 g, 61%).


3-{Ethoxycarbonylmethyl-[6-(9H-fluoren-9-ylmethoxycarbonyl-amino)-hexanoyl]-amino}-propionic acid ethyl ester AB



embedded image


Fmoc-6-amino-hexanoic acid (9.12 g, 25.83 mmol) was dissolved in dichloromethane (50 mL) and cooled with ice. Diisopropylcarbodiimde (3.25 g, 3.99 mL, 25.83 mmol) was added to the solution at 0° C. It was then followed by the addition of Diethyl-azabutane-1,4-dicarboxylate (5 g, 24.6 mmol) and dimethylamino pyridine (0.305 g, 2.5 mmol). The solution was brought to room temperature and stirred further for 6 h. Completion of the reaction was ascertained by TLC. The reaction mixture was concentrated under vacuum and ethyl acetate was added to precipitate diisopropyl area. The suspension was filtered. The filtrate was washed with 5% aqueous hydrochloric acid, 5% sodium bicarbonate and water. The combined organic layer was dried over sodium sulfate and concentrated to give the crude product which was purified by column chromatography (50% EtOAC/Hexanes) to yield 11.87 g (88%) of AB.


3-[(6-Amino-hexanoyl)-ethoxycarbonylmethyl-amino]-propionic acid ethyl ester AC



embedded image


3-{Ethoxycarbonylmethyl-[6-(9H-fluoren-9-ylmethoxycarbonylamino)-hexanoyl]-amino}-propionic acid ethyl ester AB (11.5 g, 21.3 mmol) was dissolved in 20% piperidine in dimethylformamide at 0° C. The solution was continued stirring for 1 h. The reaction mixture was concentrated under vacuum, water was added to the residue, and the product was extracted with ethyl acetate. The crude product was purified by conversion into its hydrochloride salt.


3-({6-[17-1,5-Dimethyl-hexyl)-10,13-dimethyl-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-3-yloxycarbonylamino]-hexanoyl} ethoxycarbonylmethyl-amino)-propionic acid ethyl ester AD



embedded image


The hydrdochloride salt of 3-[(6-Amino-hexanoyl)-ethoxycarbonylmethyl-amino]-propionic acid ethyl ester AC(4.7 g, 14.8 mmol) was taken up in dichloromethane. The suspension was cooled to 0° C. on ice. To the suspension diisopropylethylamine (3.87 g, 5.2 mL, 30 mmol) was added. To the resulting solution cholesteryl chloroformate (6.675 g, 14.8 mmol) was added. The reaction mixture was stirred overnight. The reaction mixture was diluted with dichloromethane and washed with 10% hydrochloric acid. The product was purified by flash chromatography (10.3 g, 92%).


1-{6-[17-(1,5-Dimethyl-hexyl)-10,13-dimethyl-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopents[a] phenanthren-3-yloxycarbonylamino]-hexanoyl}-4-oxo-pyrrolidine-3-carboxylic acid ethyl ester AE



embedded image


Potassium t-butoxide (1.1 g, 9.8 mmol) was slurried in 30 mL of dry toluene. The mixture was cooed to 0° C. on ice and 5 g (6.6 mmol) of diester AD was added slowly with stirring within 20 mins. The temperature was kept below 5° C. during the addition. The stirring was continued for 30 mins at 0° C. and 1 mL of glacial acetic acid was added, immediately followed by 4 g of NaH2PO4.H2O in 40 mL of water. The resultant mixture was extracted twice with 100 mL of dichloromethane each and the combined organic extracts were washed twice with 10 mL of phosphate buffer each, dried, and evaporated to dryness. The residue was dissolved in 60 mL of toluene, cooled to 0° C. and extracted with three 50 mL portions of cold pH 9.5 carbonate buffer. The aqueous extracts were adjusted to pH 3 with phosphoric acid, and extracted with five 40 mL portions of chloroform which were combined, dried and evaporated to dryness. The residue was purified by column chromatography using 25% ethylacetate/hexane to afford 1.9 g of b-detoester (39%).


[6-(3-Hydroxy-4-hydroxymethyl-pyrrolidin-1-yl)-6-oxo-hexyl]-carbamic acid 17-(1,5-dimethyl-hexyl)-10,13-dimethyl-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-3-yl ester AF



embedded image


Methanol (2.) was added dropwise over a period of 1 h to a refluxing mixture of b-ketoester AE (1.5 g, 2.2 mmol) and sodium borohydride (0.226 g, 6 mmol) in tetrahydrofuran (10 mL). Stirring was continued at reflux temperature for 1 h. After cooling to room temperature, 1 N NCl (12.5 mL) was added, the mixture was extracted with ethylacetate (3×40 mL). The combined ethylacetate layer was dried over anhydrous sodium sulfate and concentrated under vacuum to yield the product which was purified by column chromatography (10% MeOH/CHCl3) (89%).


(6-{3-[Bis-(4-methoxy-phenyl)-phenyl-methoxymethyl]-4-hydroxy-pyrrolidin-1-yl}-6-oxo-hexyl)-carbamic acid 17-(1,5-dimethyl-hexyl)-10,13-dimethyl-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-3-yl ester AG



embedded image


Diol AF (1.25 g, 1.994 mmol) was dried by evaporating with pyridine (2×5 mL) in vacuo. Anhydrous pyridine (10 mL) and 4,4′-dimethoxytritylchloride (0.724 g, 2.13 mmol) were added with stirring. The reaction was carried out at room temperature overnight. The reaction was quenched by the addition of methanol. The reaction mixture was concentrated under vacuum and to the residue dichloromethane (50 mL) was added. The organic layer was washed with 1 M aqueous sodium bicarbonate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated. The residual pyridine was removed by evaporating with toluene. The crude product was purified by column chromatography (2% MeOH/Chloroform, Rf=0.5 in 5% MeOH/CHCl3) (1.75 g, 95%).


Succinic acid mono-(4-[bos-(4-methoxy-phenyl-methoxymethyl]-1-{6-[17-(1,5-dimethyl-hexyl)-10,13-dimethyl 2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H cyclopenta[a]phenanthren-3-yloxycarbonylamino]-hexanoyl}-pyrrolidin-3-yl) ester AH



embedded image


Compound AG (1.0 g, 1.05 mmol) was mixed with succinic anhydride (0.150 g, 1.5 mmol) and DMAP (0.073 g, 0.6 mmol) and dried in a vacuum at 40° C. overnight. The mixture was dissolved in anhydrous dichloroethane (3 mL), triethylamine (0.318 g, 0.0440 mL, 3.15 mmol) was added and the solution was stirred at room temperature under argon atmosphere for 16 h. It was then diluted with dichloromethane (40 mL) and washed with ice cold aqueous citric acid (5 wt %, 30 mL) and water (2×20 mL). The organic phase was dried over anhydrous sodium sulfate and concentrated to dryness. The residue was used as such for the next step.


Cholesterol Derivatised CPG AI



embedded image


Succinate AH (0.254 g, 0.242 mmol) was dissolved in a mixture of dichloromethane/acetonitrile (3:2, 3 mL). To that solution DMAP (0.0296 g, 0.242 mmol) in acetonitrile (1.25 mL), 2,2′-Dithio-bis(5-nitropyridine) (0.075 g, 0.242 mmol) in acetonitrile/dichloroethane (3:1, 1.25 mL) were added successively. To the resulting solution triphenylphosphine (0.064 g, 0.242 mmol) in acetonitrile (0.6 ml) was added. The reaction mixture turned bright orange in color. The solution was agitated briefly using a wrist-action shaker (5 mins). Long chain alkyl amine-CPG (LCAA-CPG) (1.5 g, 61 mM) was added. The suspension was agitated for 2 h. The CPG was filtered through a sintered funnel and washed with acetonitrile, dichloromethane and ether successively. Unreacted amino groups were masked using acetic anhydride/hyridine. The achieved loading of the CPG was measured by taking UV measurement (37 mM/g).


The synthesis of siRNAs bearing a 5′-12-dodecanoic acid bisdecylamide group (herein referred to as “5′-C32-”) or a 5′-cholesteryl derivative group (herein referred to as “5′-Chol-”) was performed as described in WO 2004/065601, except that, for the cholesteryl derivative, the oxidation step was performed using the Beaucage reagent in order to introduce a phosphorothioate linkage at the 5′-end of the nucleic acid oligomer.


Nucleic acid sequences are represented below using standard nomenclature, and specifically the abbreviations of Table 4.









TABLE 4







Abbreviations of nucleotide monomers used in nucleic acid sequence


representation. It will be understood that these monomers, when present


in an oligonucleotide, are mutually linked by 5′-3′-phosphodiester bonds.








Abbreviation3
Nucleotide (s)





A, a
2′-deoxy-adenosine-5′-phosphate, adenosine-5′-



phosphate


C, c
2′-deoxy-cytidine-5′-phosphate, cytidine-5′-



phosphate


G, g
2′-deoxy-guanosino-5′-phosphate, guanosine-5′-



phosphate


T, t
2′-deoxy-thymidine-5′-phosphate, thymidine-5′-



phosphate


U, u
2′-deoxy-uridine-5′-phosphate, uridine-5′-phosphate


N, n
any 2′-deoxy-nucleotide/nucleotide



(G, A, C, or T, g, a, c or u)


Am
2′-O-methyladenosine-5′-phosphate


Cm
2′-O-methylcytidine phosphate


Gm
2′-O-methylguanosine-5′-phosphate


Tm
2′-O-methyl-thymidine-5′-phosphate


Um
2′-O-methyluridine-5′-phosphate


Af
2′-fluoro-2′-deoxy-adenosine-5′-phosphate


Cf
2′-fluoro-2′-deoxy-cytidine-5′-phosphate


Gf
2′-fluoro-2′-deoxy-guanosine-5′-phosphate


Tf
2′-fluoro-2′-deoxy-thymidine-5′-phosphate


Uf
2′-fluoro-2′-deoxy-uridine-5′-phosphate


A, C, G, T, U,
underlined: nucleoside-5′-phosphorothioate


a, c, g, t, u


am, cm, gm, tm,
underlined: 2-O-methyl-nucleoside-5′-phosphorothioate


um






3capital letters represent 2′-deoxribonucletides (DNA), lower case letters represent ribonucleotides (RNA)







dsRNA Expression Vectors


In another aspect of the invention , Eg5 specific dsRNA molecules that modulate Eg5 gene expression activity are expressed from transcription units inserted into DNA or RNA vectors (see, e.g., Couture, A, et al., EIG. (1996), 12:5-10; Skillern, A., et al., International PCT Publication No. WO 00/22113, Conrad, International PCT Publicatoin No. WO 00/22114, and Conrad, U.S. Pat. No. 6,054,299). These transgenes can be introduced as a linear construct, a circular plasmid, or a viral vector, which can be incorporated and inherited as a transgene integrated into the host genome. The transgene can also be constructed to permit it to be inherited as an extrachromosomal plasmid (Gassmann, et al., Proc. Natl. Acad. Sci. USA (1995) 92:1292).


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 plasmid. In preferred embodiment, a dsRNA is expressed as an inverted repeat joined by a linker polynucleotide sequence such that the dsRNA has a stem and loop structure.


The recombination dsRNA expression vectors are generally DNA plasmids or viral vectors. dsRNA expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus (for a review, see Muzyezka, et al., Curr. Topics Micro. Immunol. (1992) 158:97-129)); adenovirus (see, for example, Berkner, et al., BioTechniques (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 been used to introduce a viariety of genes into many different cell types, including epithelial cells, in vitro and/or in vivo and/or in vivo (see, e.g., Eglitis, et al., Science (1985) 230:1395-1398; Danos and Mulligan, Proc. Natl. Acad. Sci. USA (1998) 85:6460-6464; Wilson et al., 1988, Proc. Natl. Acad. Sci. USA 85:3014-3018; Armentano et al., 1990, Proc. NatI. Acad. Sci. USA 87:61416145; Huber et al., 1991, Proc. NatI. Acad. Sci. USA 88:8039-8043; Ferry et al., 1991, Proc. NatI. Acad. Sci. USA 88:8377-8381; Chowdhury et al., 1991, Science 354:1802-1805; van Beusechem. et al., 1992, Proc. Nad. Acad. Sci. USA 89:7640-19; Kay et al., 1992, Human Gene Therapy 3:641-647; Dai et al., 1992, Proc. Natl. Acad. Sci. USA 89:10892-10895; Hwu et al., 1993, J. Ummunol. 150:4104-4115; U.S. Pat. No. 4,868,116; U.S. Pat. No. 4,980,286; PCT Application WO 89/07136; PCT Application WO 89/02468; PCT Application WO 89/05344; and PCT Application WO 92/07573). Recombinant retroviral vectors capable of transducing and expressing genes inserted into the genome of a cell can be produced by transfecting the recombinant retroviral genome into suitable packaging cell lines such as PA317 and Psi-CRIP (Comette et al., 1991, Human Gene Therapy 2:5-10; Cone et al., 1984, 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. Invectious Disease, 166:769), and also have the advantage of not requiring mitotically active cells for infection.


The promoter driving dsRNA expression in either a DNA plasmid or viral vector of the invention may be a eukaryotic RNA polymerase I (e.g. ribosomal RNA promoter), RNA polymerase II (e.g. CNV early promoter or actin promoter or U1 snRNA promoter) or generally RNA polymerase III prometer (e.g. U6 snRNA or 7SK RNA promoter) or a prokaryotic promoter, for example the T7 promoter, provided the expression plasmid 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., 1986, Proc, Natl. Acad. Sci. USA 83:2511-2515)).


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 sensitive to certain physiological regulators, e.g., circulating glucose levels, or hormones (Docherty et al., 1994, FASWV J. 8:20-24). Such inducible expression systems, suitable for the control of transgene expression in cells or in mammals include regulation by ecdysone, by estrogen, progesterone, tetracycline, chemical inducers of dierization, and isopropyl-beta-D1-thiogalactopyranoside (EPTG). A person skilled in the art would be able to choose the appropriate regulatory/promoter sequence based on the intended use of the dsRNA transgene.


Generally, recombinant vectors capable of expressing dsRNA 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 reintroduction into the patient, or by any other means that allows for introduction into a desired target cell.


dsRNA expression DNA plasmids are typically transfected into target cells as a complex with cationic lipid carriers (e.g., Oligofectamine) or non-cationic lipid-based carriers (e.g. Transit-TKO™). Multiple lipid transfections for dsRNA-mediated knockdowns targeting different regions of a single Eg5 gene or multiple Eg5 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 flourescent marker, such as Green Fluorescent Protein (GFP). Stable transfection of ex vivo cells can be ensured using markers that provide the transfected cell with resistance to specific environmental factors (e.g., antibiotics and drugs), such as hygromycin B resistance.


The Eg5 dsRNA molecules can also be inserted into vectors and sued 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. Pat. No. 5,328,470) or by stereotactic injection (see e.g., Chen et al. (1994) Proc. Natl. Acad. Sci. USA 91:3045-3057). 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 delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene delivery system.


Eg5 siRNA in vitro Screening via Cell Proliferation


As silencing of Eg5 has been shown to cause mitotic arrest (Weil, D, et al [2002] Biotechniques 33: 1244-8), a cell viability assay was used for siRNA activity screening. HeLa cells (14000 per well [Screens 1 and 3] or 10000 per well [Screen2])) were seeded in 96-well plates and simultaneously transfected with Lipofectamine 2000 (Invitrogen) at a final siRNA concentration in the well of 30 nM and at final concentrations of 50 nM (1st screen) and 25 nM (2nd screen). A subset of duplexes was tested at 25 nM in a third screen (Table 5).


Seventy-two hours post-transfection, cell proliferation was assayed the addition of WST-1 reagent (Roche) to the culture medium, and subsequent absorbance measurement at 450 nm. The absorbance value for control (non-transfected) cells was considered 100 percent, and absorbances for the siRNA transfected wells were compared to the control value. Assays were performed in sextuplicate for each of three screens. A subset of the siRNAs was further tested at a range of siRNA concentrations. Assays were performed in HeLa cells (14000 per well; method same as above, Table 5).












TABLE 5









Relative absorbance




at 450 nm














Screen I

Screen II

Screen III



Duplex
mean
sd
Mean
sd
mean
Sd
















AL-DP-6226
20
10
28
11
43
9


AL-DP-6227
66
27
96
41
108
33


AL-DP-6228
56
28
76
22
78
18


AL-DP-6229
17
3
31
9
48
13


AL-DP-6230
48
8
75
11
73
7


AL-DP-6231
8
1
21
4
41
10


AL-DP-6232
16
2
37
7
52
14


AL-DP-6233
31
9
37
6
49
12


AL-DP-6234
103
40
141
29
164
45


AL-DP-6235
107
34
140
27
195
75


AL-DP-6236
48
12
54
12
56
12


AL-DP-6237
73
14
108
18
154
37


AL-DP-6238
64
9
103
10
105
24


AL-DP-6239
9
1
20
4
31
11


AL-DP-6240
99
7
139
16
194
43


AL-DP-6241
43
9
54
12
66
19


AL-DP-6242
6
1
15
7
36
8


AL-DP-6243
7
2
19
5
33
13


AL-DP-6244
7
2
19
3
37
13


AL-DP-6245
25
4
45
10
58
9


AL-DP-6246
34
8
65
10
66
13


AL-DP-6247
53
6
78
14
105
20


AL-DP-6248
7
0
22
7
39
12


AL-DP-6249
36
8
48
13
61
7









The nine siRNA duplexes that showed the greatest growth inhibition in Table 5 were re-tested at a range of siRNA concentrations in HeLa cells. The siRNA concentrations tested were 100 nM, 33.3 nM, 11.1 nM, 3.70 nM, 1.23 nM, 0.41 nM and 0.046 nM. Assays were performed in sextuplicate, and the concentration of each siRNA resulting in fifty percent inhibition of cell proliferation (IC50) was calculated. This dose-response analysis was performed between two and four times for each duplex. Mean IC50 values (nM) are given in Table 6.












TABLE 6







Duplex
Mean IC50



















AL-DP-6226
15.5



AL-DP-6229
3.4



AL-DP-6231
4.2



AL-DP-6232
17.5



AL-DP-6239
4.4



AL-DP-6242
5.2



AL-DP-6243
2.6



AL-DP-6244
8.3



AL-DP-6248
1.9










Eg5 siRNA in vitro Screening via Cell Proliferation


Directly before transfection, Hela S3 (ATCC-Number: CCL-2:2, LCG Promochem GmbH, Wesel, Germany) cells were seeded at 1.5×104 cells/well on 96-well plates (Greiner Bio-One GmbH, Frickenhausen, Germany) in 75 μl of growth medium (Ham's F12, 10% fetal calf serum, 100 u penicillin/100 μg/ml streptomycin, all from Biochrom AG, Berlin, Germany). Transfections were performed in quadruplicates. For each well 0.5 μl Lipofectamine2000 (Invitrogen GmbH, Darlsruhe, Germany) were mixed with 12 μl Opti-MEM (Invitrogen) and incubated for 15 min at room temperature. For the siRNA concentration being 50 nM in the 100 μl transfection volume, 1 μl of a 5 μM siRNA were mixed with 11.5 μl Opti-MEM per well, combined with the Lipofectamine2000-Opti-MEM mixture and again incubated for 15 minutes at room temperature, siRNA-Lipofectamine2000-complexes were applied completely (25 μl each per well) to the cells and cells were incubated for 24 h at 37° C. and 5% CO2 in a humidified incubator (Heracus GmbH, Hanau). The single dose screen was done once at 50 nM and at 25 nM, respectively.


Cells were harvested by applying 50 μl of lysis mixture (content of the QuantiGene bDNA-kit from Genospectra, Fremont, USA) to each well containing 100 μl of growth medium and were lysed at 53° C. for 30 min. Afterwards, 50 μl of the lysates were incubated with probesets specific to human Eg5 and human GAPDH and proceeded according to the manufacturer's protocol for QuantiGene. In the end chemoluminescence was measured in a Victor2-Light (Perkin Elmer, Wiesbaden, Germany) as RLUs (relative light units) and values obtained with the hEg5 probeset were normalized to the respective GAPDH values for each well. Values obtained with siRNAs directed against Eg5 were related to the value obtained with an unspecific siRNA (directed against HCV) which was set to 100% (Tables 1, 2, and 3).


Effective siRNAs from the screen were further characterized by dose response curves. Transfections of dose response curves were performed at the following concentrations: 100 nM, 16.7 nM, 2.8 nM, 0.46 nM, 77 picoM, 12.8 picoM, 2.1 picoM, 0.35 picoM, 59.5 fM, 9.9 fM and mock (no siRNA) and diluted with Opti-MEM to a final concentration of 12.5 μl according to the above protocol. Data analysis was performed by using the Microsoft Excel add-in software XL-fit 4.2 (IDBS, Guildford, Surrey, UK) and applying the dose response model number 205 (Tables 1, 2 and 3).


The lead siRNA AD12115 was additionally analyzed by applying the WST-proliferation assay from Roche (as previously described).


A subset of 34 duplexes from Table 2 that showed greatest activity was assayed by transfection in HeLa cells at final concentrations ranging from 100 nM to 10 fM. Transfections were performed in quadruplicate. Two dose-response assays were performed for each duplex. The concentration giving 20% (IC20), 50% (IC50) and 80% (IC80) reduction of KSP mRNA was calculated for each duplex. (Table 7).









TABLE 7







Concentrations given in pM














IC20s

IC50s

IC80s



Duplex name
1st screen
2nd screen
1st screen
2nd screen
1st screen
2nd screen
















AD12077
1.19
0.80
6.14
10.16
38.36
76.16


AD12078
25.43
25.43
156.18
156.18
ND
ND


AD12085
9.08
1.24
40.57
8.52
257.68
81.26


AD12095
1.03
0.97
9.84
4.94
90.31
60.47


AD12113
4.00
5.94
17.18
28.14
490.83
441.30


AD12115
0.60
0.41
3.79
3.39
23.45
23.45


AD12125
31.21
22.02
184.28
166.15
896.85
1008.11


AD12134
2.59
5.51
17.87
22.00
116.36
107.03


AD12149
0.72
0.50
4.51
3.91
30.29
40.89


AD12151
0.53
6.84
4.27
10.72
22.88
43.01


AD12152
155.45
7.56
867.36
66.69
13165.27
ND


AD12157
0.30
26.23
14.60
92.08
14399.22
693.31


AD12166
0.20
0.93
3.71
3.86
46.28
20.59


AD12180
28.85
28.85
101.06
101.06
847.21
847.21


AD12185
2.60
0.42
15.55
13.91
109.80
120.63


AD12194
2.08
1.11
5.37
5.09
53.03
30.92


AD12211
5.27
4.52
11.73
18.93
26.74
191.07


AD12257
4.56
5.20
21.68
22.75
124.69
135.82


AD12280
2.37
4.53
6.89
20.23
64.80
104.82


AD12281
8.81
8.65
19.68
42.89
119.01
356.08


AD12282
7.71
456.42
20.09
558.00
ND
ND


AD12285
ND
1.28
57.30
7.31
261.79
42.53


AD12292
40.23
12.00
929.11
109.10
ND
ND


AD12252
0.02
18.63
6.35
68.24
138.09
404.91


AD12275
25.76
25.04
123.89
133.10
1054.54
776.25


AD12266
4.85
7.80
10.00
32.94
41.67
162.65


AD12267
1.39
1.21
12.00
4.67
283.03
51.12


AD12264
0.92
2.07
8.56
15.12
56.36
196.78


AD12268
2.29
3.67
22.16
25.64
258.27
150.84


AD12279
1.11
28.54
23.19
96.87
327.28
607.27


AD12256
7.20
33.52
46.49
138.04
775.54
1076.76


AD12259
2.16
8.31
8.96
40.12
50.05
219.42


AD12276
19.49
6.14
89.60
59.60
672.51
736.72


AD12321
4.67
4.91
24.88
19.43
139.50
89.49





ND-not determined






Silencing of Liver Eg5/KSP in Juvenile Rats Following Single-bolus Administration of LNP01 Formulated siRNA


From birth until approximately 23 days of age, Eg5/KSP expression can be detected in the growing rat liver. Target silencing with a formulated Eg5/KSP siRNA was evaluated in juvenile rats.


KSP Duplex Tested















Duplex ID
Target
Sense
Antisense



















AD6248
Eg5
AccGAAGuGuuGuuuGuccTsT
GGAcAAAcAAcACUUCGGUTsT





(SEQ ID NO:1238)
(SEQ ID NO:1239)









Methods


Dosing of animals. Male, juvenile Sprague-Dawley rats (19 days old) were administered single doses of lipidoid (“LNP01”) formulated siRNA via tail vein injection. Groups of ten animals received doses of 10 milligrams per kilogram (mg/kg) bodyweight of either AD6248 or an unspecific siRNA. Dose level refers to the amount of siRNA duplex administered in the formulation. A third group received phosphate-buffered saline. Animals were sacrificed two days after siRNA administration. Livers were dissected, flash frozen in liquid Nitrogen and pulverized into powders.


mRNA measurements. Levels of Eg5/KSP mRNA were measured in livers from all treatment groups. Samples of each liver powder (approximately ten milligrams) were homogenized in tissue lysis buffer containing proteinase K. Levels of Eg5/KSP and GAPDH mRNA were measured in triplicate for each sample using the Quantigene branched DNA assay (GenoSpectra). Mean values for Eg5/KSP were normalized to mean GAPDH values for each sample. Group means were determined and normalized to the PBS group for each experiment.


Statistical analysis. Significance was determined by ANOVA followed by the Tukey post-hoc test


Results


Data Summary


Mean values (±standard deviation) for Eg5/KSP mRNA are given. Statistical significance (p value) versus the PBS group is shown (ns, not significant [p>0.05]).


Experiment 1















VEGF/GAPDH
p value





















PBS

 1.0 ± 0.47




AD6248
10 mg/kg
0.47 ± 0.12
<0.001



unspec
10 mg/kg
 1.0 ± 0.26
ns










A statistically significant reduction in liver Eg5/KSP mRNA was obtained following treatment with formulated AD6248 at a dose of 10 mg/kg.


Silencing of Rat Liver VEGF Following Intravenous Infusion of LNP01 Formulated siRNA Duplexes


A “lipidoid” formulation comprising an equimolar mixture of two siRNAs was administered to rats. One siRNA (AD3133) was directed towards VEGF. The other (AD12115) was directed towards Eg5/KSP. Since Eg5/KSP expression is nearly undetectable in the adult rat liver, only VEGF levels were measured following siRNA treatment.


siRNA Duplexes Administered















Duplex ID
Target
Sense
Antisense



















AD12115
Eg5/KSP
ucGAGAAucuAAAcuAAcuTsT
AGUuAGUUuAGAUUCUCGATsT





(SEQ ID NO:1240)
(SEQ ID NO:1241)





AD3133
VEGF
GcAcAuAGGAGAGAuGAGCUsU
AAGCUcAUCUCUCCuAuGuG




(SEQ ID NO:1242)
CusG





(SEQ ID NO:1243)





Key: A, G, C, U - ribonucleotides; c, u - 2′-O-Me ribonucleotides; s-phosphorothioate.






Methods


Dosing of animals. Adult, female Sprague-Dawley rats were administered lipidoid (“LNP01”) formulated siRNA by a two-hour infusion into the femoral vein. Groups of four animals received doses of 5, 10 and 15 milligrams per kilogram (mg/kg) bodyweight of formulated siRNA. Dose level refers to the total amount of siRNA duplex administered in the formulation. A fourth group received phosphate-buffered saline. Animals were sacrificed 72 hours after the end of the siRNA infusion. Livers were dissected, flash frozen in liquid Nitrogen and pulverized into powders.


Formulation Procedure


The lipidoid ND98-4HCl (MW 1487) (Formula 1), Cholesterol (Sigma-Aldrich), and PEG-Ceramide C16 (Avanti Polar Lipids) were used to prepare lipid-siRNA nanoparticles. Stock solutions of each in ethanol were prepared: ND98, 133 mg/mL; Cholesterol, 25 mg/mL, PEG-Ceramide C16, 100 mg/mL. ND98, Cholesterol, and PEG-Ceramide C16 stock solutions were then combined in a 42:48:10 molar ratio. Combined lipid solution was mixed rapidly with aqueous siRNA (in sodium acetate pH 5) such that the final ethanol concentration was 35-45% and the final sodium acetate concentration was 100-300 mM. Lipid-siRNA nanoparticles formed spontaneously upon mixing. Depending on the desired particle size distribution, the resultant nanoparticle mixture was in some cases extruded through a polycarbonate membrane (100 nm cut-off) using a thermobarrel extruder (Lipex Extruder, Northern Lipids, Inc). In other cases, the extrusion step was omitted. Ethanol removal and simultaneous buffer exchange was accomplished by either dialysis or tangential flow filtration. Buffer was exchanged to phosphate buffered saline (PBS) pH 7.2.




embedded image


Characterization of Formulations


Formulations prepared by either the standard or extrusion-free method are characterized in a similar manner. Formulations are first characterized by visual inspection. They should be whitish translucent solutions free from aggregates or sediment. Particle size and particle size distribution of lipid-nanoparticles are measured by dynamic light scattering using a Malvern Zetasizer Nano ZS (Malvern, USA). Particles should be 20-300 nm, and ideally, 40-100 nm in size. The particle size distribution should be unimodal. The total siRNA concentration in the formulation, as well as the entrapped fraction, is estimated using a dye exclusion assay. A sample of the formulated siRNA is incubated with the RNA-binding dye Ribogreen (Molecular Probes) in the presence or absence of a formulation disrupting surfactant, 0.5% Triton-X100. The total siRNA in the formulation is determined by the signal from the sample containing the surfactant, relative to a standard curve. The entrapped fraction is determined by subtracting the “free” siRNA content (as measured by the signal in the absence of surfactant) from the total siRNA content. Percent entrapped siRNA is typically >85%.


mRNA measurements. Samples of each liver powder (approximately ten milligrams) were homogenized in tissue lysis buffer containing proteinase K. Levels of VEGF and GAPDH mRNA were measured in triplicate for each sample using the Quantigene branched DNA assay (GenoSpectra). Mean values for VEGF were normalized to mean GAPDH values for each sample. Group means were determined and normalized to the PBS group for each experiment.


Protein measurements. Samples of each liver powder (approximately 60 milligrams) were homogenized in 1 ml RIPA buffer. Total protein concentrations were determined using the MicroBCA protein assay kit (Pierce). Samples of total protein from each animal was used to determine VEGF protein levels using a VEGF ELISA assay (R&D systems). Group means were determined and normalized to the PBS group for each experiment.


Statistical analysis. Significance was determined by ANOVA followed by the Tukey post-hoc test


Results


Data Summary


Mean values (±standard deviation) for mRNA (VEGF/GAPDH) and protein (rel. VEGF) are shown for each treatment group. Statistical significance (p value) versus the PBS group for each experiment is shown.


















VEGF/GAPDH
p value
rel VEGF
p value




















PBS
 1.0 ± 0.17

 1.0 ± 0.17



 5 mg/kg
0.74 ± 0.12
<0.05
0.23 ± 0.03
<0.001


10 mg/kg
0.65 ± 0.12
<0.005
0.22 ± 0.03
<0.001


15 mg/kg
0.49 ± 0.17
<0.001
0.20 ± 0.04
<0.001









Statistically significant reductions in liver VEGF mRNA and protein were measured at all three siRNA dose levels.












TABLE 1








sequence





of total




position
23 mer




in human
target




access. #
site
SEQ I
sense sequence (5′-3′)













385-407
ACCGAAGUGUUGUUUGUCCAAUU
1
cGAAGuGuuGuuuGuccAATsT


347-369
UAUGGUGUUUGGAGCAUCUACUA
3
uGGuGuuuGGAGcAucuAcTsT


1078-1100
AAUCUAAACUAACUAGAAUCCUC
5
ucuAAAcuAAcuAGAAuccTsT


1067-1089
UCCUUAUCGAGAAUCUAAACUAA
7
cuuAucGAGAAucuAAAcuTsT


374-396
GAUUGAUGUUUACCGAAGUGUUG
9
uuGAuGuuuAccGAAGuGuTsT


205-227
UGGUGAGAUGCAGACCAUUUAAU
11
GuGAGAuGcAGAccAuuuATsT


1176-1198
ACUCUGAGUACAUUGGAAUAUGC
13
ucuGAGuAcAuuGGAAuAuTsT


386-408
CCGAAGUGUUGUUUGUCCAAUUC
15
GAAGuGuuGuuuGuccAAuTsT


416-438
AGUUAUUAUGGGCUAUAAUUGCA
17
uuAuuAuGGGcuAuAAuuGTsT


485-507
GGAAGGUGAAAGGUCACCUAAUG
19
AAGGuGAAAGGucAccuAATsT


476-498
UUUUACAAUGGAAGGUGAAAGGU
21
uuAcAAuGGAAGGuGAAAGTsT


486-508
GAAGGUGAAAGGUCACCUAAUGA
23
AGGuGAAAGGucAccuAAuTsT


487-509
AAGGUGAAAGGUCACCUAAUGAA
25
GGuGAAAGGucAccuAAuGTsT


1066-1088
UUCCUUAUCGAGAAUCUAAACUA
27
ccuuAucGAGAAucuAAAcTsT


1256-1278
AGCUCUUAUUAAGGAGUAUACGG
29
cucuuAuuAAGGAGuAuAcTsT


2329-2351
CAGAGAGAUUCUGUGCUUUGGAG
31
GAGAGAuucuGuGcuuuGGTsT


1077-1099
GAAUCUAAACUAACUAGAAUCCU
33
AucuAAAcuAAcuAGAAucTsT


1244-1266
ACUCACCAAAAAAGCUCUUAUUA
35
ucAccAAAAAAGcucuuAuTsT


637-659
AAGAGCUUUUUGAUCUUCUUAAU
37
GAGcuuuuuGAucuucuuATsT


1117-1139
GGCGUACAAGAACAUCUAUAAUU
39
cGuAcAAGAAcAucuAuAATsT


373-395
AGAUUGAUGUUUACCGAAGUGUU
41
AuuGAuGuuuAccGAAGuGTsT


1079-1101
AUCUAAACUAACUAGAAUCCUCC
43
cuAAAcuAAcuAGAAuccuTsT


383-405
UUACCGAAGUGUUGUUUGUCCAA
45
AccGAAGuGuuGuuuGuccTsT


200-222
GGUGGUGGUGAGAUGCAGACCAU
47
uGGuGGuGAGAuGcAGAccTsT



















single







dose
SDs 2nd






screen @
screen


position



25 nM [%
(among


in human


duplex
residual
quadrupli-


access. #
SEQ I
antisense sequence (5′-3′)
name
mRNA]
cates)















385-407
2
UUGGAcAAAcAAcACUUCGTsT


embedded image


23%
3%


347-369
4
GuAGAUGCUCcAAAcACcATsT
AL-DP-6227
69%
10%


1078-1100
6
GGAUUCuAGUuAGUUuAGATsT
AL-DP-6228
33%
2%


1067-1089
8
AGUUuAGAUUCUCGAuAAGTsT


embedded image


2%
2%


374-396
10
AcACUUCGGuAAAcAUcAATsT
AL-DP-6230
66%
11%


205-227
12
uAAAUGGUCUGcAUCUcACTsT


embedded image


17%
1%


1176-1198
14
AuAUUCcAAUGuACUcAGATsT


embedded image


9%
3%


386-408
16
AUUGGAcAAAcAAcACUUCTsT
AL-DP-6233
24%
6%


416-438
18
cAAUuAuAGCCcAuAAuAATsT
AL-DP-6234
91%
2%


485-507
20
UuAGGUGACCUUUcACCUUTsT
AL-DP-6235
112%
4%


476-498
22
CUUUcACCUUCcAUUGuAATsT
AL-DP-6236
69%
4%


486-508
24
AUuAGGUGACCUUUcACCUTsT
AL-DP-6237
42%
2%


487-509
26
cAUuAGGUGACCUUUcACCTsT
AL-DP-6238
45%
2%


1066-1088
28
GUUuAGAUUCUCGAuAAGGTsT


embedded image


2%
1%


1256-1278
30
GuAuACUCCUuAAuAAGAGTsT
AL-DP-6240
48%
2%


2329-2351
32
CcAAAGcAcAGAAUCUCUCTsT
AL-DP-6241
41%
2%


1077-1099
34
GAUUUuAGUuAGUUuAGAUTsT


embedded image


8%
2%


1244-1266
36
AuAAGAGCUUUUUUGGUGATsT


embedded image


7%
1%


637-659
38
uAAGAAGAUcAAAAAGCUCTsT


embedded image


6%
2%


1117-1139
40
UuAuAGAUGUUCUUGuACGTsT
AL-DP-6245
12%
2%


373-395
42
cACUUCGGuAAAcAUcAAUTsT
AL-DP-6246
28%
3%


1079-1101
44
AGGAUUCuAGUuAGUUuAGTsT
AL-DP-6247
71%
4%


383-405
46
GGAcAAAcAAcACUUCGGUTsT


embedded image


5%
2%


200-222
48
GGUCUGcAYCUcACcACcATsT
AL-DP-6249
28%
3%























TABLE 2A





position
SEQ
sequence of
SEQ

SEQ
sense
du-


in human
ID
total 19mer
ID

ID
sequence
plex


access. #
NO:
target site
NO:
sense sequence (5′-3′)
NO:
(5′-3′)
name






















 829-847
1268
CAUACUCAUGUCGUUCCCA
49
cAuAcucuAGucGuucccATsT
50
UGGGAACGACuAGAGuAUGTsT
AD-









12072





 246-264
1269
AGCGCCCAUUCAAUAGUA-
51
AGcGcccAuucAAuAGuAGTsT
52
CuACuAUUGAAUGGGCGCUTsT
AD-




G




12073





 238-256
1270
GGAAAGCUAGCGCCCAUUC
53
GGAAAGcuAGcGcccAuucTsT
54
GAAUGGGCGCuAGCUUUCCTsT
AD-









12074





 239-257
1271
GAAAGCUAGCGCCCAUUCA
55
GAAAGcuAGcGcccAuucATsT
56
UGAAUGGGCGCuAGCUUUCTsT
AD-









12075





 878-896
1272
AGAAACUACGAUUGAUGG-
57
AGAAAcuAcGAuuGAuGGATsT
58
UCcAUcAAUCGuAGUUUCUTsT
AD-




A




12076





1064-1082
1273
UGUUCCUUAUCGAGAAUC-
59
uGuuccuuAucGAGAAucuTsT
60
AGAUUCUCGAuAAGGAAcATsT
AD-




U




12077





3278-3296
1274
CAGAUUACCUCUGCGAGCC
61
cAGAuuAccucuGcGAGccTsT
62
GGCUCGcAGAGGuAAUCUGTsT
AD-









12078





 247-265
1275
GCGCCCAUUCAAUAGUAG-
63
GcGcccAuucAAuAGuAGATsT
64
UCuACuAUUGAAUGGGCGCTsT
AD-




A




12079





 434-452
1276
UUGCACUAUCUUUGCGUA-
65
uuGcAcuAucuuuGcGuAuTsT
66
AuACGcAAAGAuAGUGcAATsT
AD-




U




12080





 232-250
1277
CAGAGCGGAAAGCUAGCG-
67
cAGAGcGGAAAGcuAGcGcTsT
68
GCGCuAGCUUUCCGCUCUGTsT
AD-




C




12081





1831-1849
1278
AGACCUUAUUUGGUAAUC-
69
AGAccuuAuuuGGuAAucuTsT
70
AGAUuACcAAAuAAGGUCUTsT
AD-




U




12082





1105-1123
1279
AUUCUCUUGGAGGGCGUA-
71
AuucucuuGGAGGGcGuAcTsT
72
GuACGCCCUCcAAGAGAAUTsT
AD-




C




12083





 536-554
1280
GGCUGGUAUAAUUCCACG-
73
GGcuGGuAuAAuuccAcGuTsT
74
ACGUGGAAUuAuACcAGCCTsT
AD-




U




12084





 236-254
1281
GCGGAAAGCUAGCGCCCAU
75
GcGGAAAGcuAGcGcccAuTsT
76
AUGGGCGCuAGCUUUCCGCTsT
AD-









12085





 435-453
1282
UGCACUAUCUUUGCGUAU-
77
uGcAcuAucuuuGcGuAuGTsT
78
cAuACGcAAAGAuAGUGcATsT
AD-




G




12086





 541-559
1283
GUAUAAUUCCACGUACCCU
79
GuAuAAuuccAcGuAcccuTsT
80
AGGGuACGUGGAAUuAuACTsT
AD-









12087





1076-1094
1284
AGAAUCUAAACUAACUAG-
81
AGAAucuAAAcuAAcuAGATsT
82
UCuAGUuAGUUuAGAUUCUTsT
AD-




A




12088





1432-1450
1285
AGGAGCUGAAUAGGGUUA-
83
AGGAGcuGAAuAGGGuuAcTsT
84
GuAACCCuAUUcAGCUCCUTsT
AD-




C




12089





1821-1839
1286
GAAGUACAUAAGACCUUA-
85
GAAGuAcAuAAGAccuuAuTsT
86
AuAAGGUCUuAUGuACUUCTsT
AD-




U




12090





2126-2144
1287
GACAGUGGCCGAUAAGAU-
87
GAcAGuGGccGAuAAGAuATsT
88
uAUCUuAUCGGCcACUGUCTsT
AD-




A




12091





2373-2391
1288
AAACCACUUAGUAGUGUC-
89
AAAccAcuuAGuAGuGuccTsT
90
GGAcACuACuAAGUGGUUUTsT
AD-




C




12092





4026-4044
1289
UCCCUAGACUUCCCUAUUU
91
ucccuAGAcuucccuAuuuTsT
92
AAAuAGGGAAGUCuAGGGATsT
AD-









12093





4030-4048
1290
UAGACUUCCCUAUUUCGCU
93
uAGAcuucccuAuuucGcuTsT
94
AGCGAAAuAGGGAAGUCuATsT
AD-









12094





 144-162
1291
GCGUCGCAGCCAAAUUCGU
95
GcGucGcAGccAAAuucGuTsT
96
ACGAAUUUGGCUGCGACGCTsT
AD-









12095





 242-260
1292
AGCUAGCGCCCAUUCAAUA
97
AGcuAGcGcccAuucAAuATsT
98
uAUUGAAUGGGCGCuAGCUTsT
AD-









12096





 879-897
1293
GAAACUACGAUUGAUGGA-
99
GAAAcuAcGAuuGAuGGAGTsT
100
CUCcAUcAAUCGuAGUUUCTsT
AD-




G




12097





2134-2152
1294
CCGAUAAGAUAGAAGAUC-
101
ccGAuAAGAuAGAAGAucATsT
102
UGAUCUUCuAUCUuAUCGGTsT
AD-




A




12098





 245-263
1295
UAGCGCCCAUUCAAUAGU-
103
uAGcGcccAuucAAuAGuATsT
104
uACuAUUGAAUGGGCGCuATsT
AD-




A




12099





 444-462
1296
UUUGCGUAUGGCCAAACU-
195
uuuGcGuAuGGccAAAcuGTsT
106
cAGUUUGGCcAuACGcAAATsT
AD-




G




12100





 550-568
1297
CACGUACCCUUCAUCAAAU
107
cAcGuAcccuucAucAAAuTsT
108
AUUUGAUGAAGGGuACGUGTsT
AD-1









12101





 442-460
1298
UCUUUGCGUAUGGCCAAA-
109
ucuuuGcGuAuGGccAAAcTsT
110
GUUUGGCcAuACGcAAAGATsT
AD-




C




12102





 386-440
1299
CCGAAGUGUUGUUUGUCC-
111
ccGAAGuGuuGuuuGuccATsT
112
UGGAcAAAcAAcACUUCGGTsT
AD-




A




12103





 233-251
1300
AGAGCGGAAAGCUAGCGC-
113
AGAGcGGAAAGcuAGcGccTsT
114
GGCGCuAGCUUUCCGCUCUTsT
AD-




C




12104





 243-261
1301
GCUAGCGCCCAUUCAAUAG
115
GcuAGcGcccAuucAAuAGTsT
116
CuAUUGAAUGGGCGCuAGCTsT
AD-









12105





 286-304
1302
AAGUUAGUGUACGAACUG-
117
AAGuuAGuGuAcGAAcuGGTsT
118
CcAGUUCGuAcACuAACUUTsT
AD-




G




12106





 294-312
1303
GUACGAACUGGAGGAUUG
119
GuAcGAAcuGGAGGAuuGGTsT
120
CcAAUCCUCcAGUUCGuACTsT
AD-









12107





 296-314
1304
ACGAACUGGAGGAUUGGC-
121
AcGAAcuGGAGGAuuGGcuTsT
122
AGCcAAUCCUCcAGUUCGUTsT
AD-




U




12108





 373-391
1305
AGAUUGAUGUUUACCGAA-
123
AGAuuGAuGuuuAccGAAGTsT
124
CUUCGGuAAAcAUcAAUCUTsT
AD-




G




12109





 422-440
1306
UAUGGGCUAUAAUUGCAC-
125
uAuGGGcuAuAAuuGcAcuTsT
126
AGUGcAAUuAuAGCCcAuATsT
AD-




U




12110





 441-459
1307
AUCUUUGCGUAUGGCCAA-
127
AucuuuGcGuAuGGccAAATsT
128
UUUGGCcAuACGcAAAGAUTsT
AD-




A




12111





 832-850
1308
ACUCUAGUCGUUCCCACUC
129
AcucuAGucGuucccAcucTsT
130
GAGUGGGAACGACuAGAGUTsT
AD-









12112





 881-899
1309
AACUACGAUUGAUGGAGA-
131
AAcuAcGAuuGAuGGAGAATsT
132
UUCUCcAUcAAUCGuAGUUTsT
AD-




A




12113





 975-993
1310
GAUAAGAGAGCUCGGGAA-
133
GAuAAGAGAGcucGGGAAGTsT
134
CUUCCCGAGCUCUCUuAUCTsT
AD-




G




12114





1073-1091
1311
UCGAGAAUCUAAACUAAC-
135
ucGAGAAucuAAAcuAAcuTsT
136
AGUuAGUUuAGAUUCUCGATsT
AD-




U




12115





1084-1102
1312
AACUAACUAGAAUCCUCCA
137
AAcuAAcuAGAAuccuccATsT
138
UGGAGGAUUCuAGUuAGUUTsT
AD-









12116





1691-1709
1313
GGAUCGUAAGAAGGCAGU-
139
GGAucGuAAGAAGGcAGuuTsT
140
AACUGCCUUCUuACGAUCCTsT
AD-




U




12117





1693-1711
1314
AUCGUAAGAAGGCAGUUG-
141
AucGuAAGAAGGcAGuuGATsT
142
UcAACUGCCUUCUuACGAUTsT
AD-




A




12118





1702-1720
1315
AGGCAGUUGACCAACACA-
143
AGGcAGuuGAccAAcAcAATsT
144
UUGUGUUGGUcAACUGCCUTsT
AD-




A




12119





2131-2149
1316
UGGCCGAUAAGAUAGAAG-
145
uGGccGAuAAGAuAGAAGATsT
146
UCUUCuAUCUuAUCGGCcATsT
AD-




A




12120





2412-2430
1317
UCUAAGGAUAUAGUCAAC-
147
ucuAAGGAuAuAGucAAcATsT
148
UGUUGACuAuAUCCUuAGATsT
AD-




A




12121





2859-2877
1318
ACUAAGCUUAAUUGCUUU-
149
AcuAAGcuuAAuuGcuuucTsT
150
GAAAGcAAUuAAGCUuAGUTsT
AD-




C




12122





3294-3312
1319
GCCCAGAUCAACCUUUAAU
151
GcccAGAucAAccuuuAAuTsT
153
AUuAAAGGUUGAUCUGGGCTsT
AD-









12123





 223-241
1320
UUAAUUGGGCAGAGCGGA-
153
uuAAuuuGGcAGAGcGGAATsT
154
UUCCGCUCUGCcAAAUuAATsT
AD-




A




12124





1070-1088
1321
UUAUCGAGAAUCUAAACU-
155
uuAucGAGAAucuAAAcuATsT
156
uAGUUuAGAUUCUCGAuAATsT
AD-




A




12125





 244-262
1322
CUAGCGCCCAUUCAAUAGU
157
cuAGcGcccAuucAAuAGuTsT
158
ACuAUUGAAUGGGCGCuAGTsT
AD-









12126





 257-275
1323
AAUAGUAGAAUGUGAUCC-
159
AAuAGuAGAAuGuGAuccuTsT
160
AGGAUcAcAUUCuACuAUUTsT
AD-




U




12127





 277-295
1324
UACGAAAAGAAGUUAGUG-
161
uAcGAAAAGAAGuuAGuGuTsT
162
AcACuAACUUCUUUUCGuATsT
AD-




U




12128





 284-302
1325
AGAAGUUAGUGUACGAAC-
163
AGAAGuuAGuGuAcGAAcuTsT
164
AGUUCGuAcACuAACUUCUTsT
AD-




U




12129





 366-384
1326
ACUAAACAGAUUGAUGUU-
165
AcuAAAcAGAuuGAuGuuuTsT
166
AAAcAUcAAUCUGUUuAGUTsT
AD-




U




12130





 443-461
1327
CUUUGCGUAUGGCCAAAC-
167
cuuuGcGuAuGGccAAAcuTsT
168
AGUUUGGCcAuACGcAAAGTsT
AD-




U




12131





 504-522
1328
AAUGAAGAGUAUACCUGG-
169
AAuGAAGAGuAuAccuGGGTsT
170
CCcAGGuAuACUCUUcAUUTsT
AD-




G




12132





 543-561
1329
AUAAUUCCACGUACCCUUC
171
AuAAuuccAcGuAcccuucTsT
172
GAAGGGuACGUGGAAUuAUTsT
AD-









12133





 551-569
1330
ACGUACCCUUCAUCAAAUU
173
AcGuAcccuucAucAAAuuTsT
174
AAUUUGAUGAAGGGuACGUTsT
AD-









12134





 552-570
1331
CGUACCCUUCAUCAAAUUU
175
cGuAcccuucAucAAAuuuTsT
176
AAAUUUGAUGAAGGGuACGTsT
AD-









12135





 553-571
1332
GUACCCUUCAUCAAAUUU-
177
GuAcccuuuAucAAAuuuuTsT
178
AAAAUUUGAUGAAGGGuACTsT
AD-




U




12136





 577-595
1333
AACUUACUGAUAAUGGUA-
179
AAcuuAcuGAuAAuGGuAcTsT
180
GuACcAUuAUcAGuAAGUUTsT
AD-




C




12137





 602-620
1334
UUCAGUCAAAGUGUCUCU-
181
uucAGucAAAGuGucucuGTsT
182
cAGAGAcACUUUGACUGAATsT
AD-




G




12138





 652-670
1335
UUCUUAAUCCAUCAUCUG-
183
uucuuAAuccAucAucuGATsT
184
UcAGAUGAUGGAUuAAGAATsT
AD-




A




12139





 747-765
1336
ACAGUACACAACAAGGAU-
185
AcAGuAcAcAAcAAGGAuGTsT
186
cAUCCUUGUUGUGuACUGUTsT
AD-




G




12140





 877-895
1337
AAGAAACUACGAUUGAUG-
187
AAGAAAcuAcGAuuGAuGGTsT
188
CcAUcAAUCGuAGUUUCUUTsT
AD-




G




12141





 880-898
1338
AAACUACGAUUGAUGGAG-
189
AAAcuAcGAuuGAuGGAGATsT
190
UCUCcAUcAAUCGuAGUUUTsT
AD-




A




12142





 965-983
1339
UGGAGCUGUUGAUAAGAG-
191
uGGAGcuGuuGAuAAGAGATsT
192
UCUCUuAUcAAcAGCUCcATsT
AD-




A




12143





1086-1104
1340
CUAACUAGAAUCCUCCAGG
193
cuAAcuAGAAuccuccAGGTsT
194
CCUGGAGGAUUCuAGUuAGTsT
AD-









12144





1191-1209
1341
GAAUAUGCUCAUAGAGCA-
195
GAAuAuGcucAuAGAGcAATsT
196
UUGCUCuAUGAGcAuAUUCTsT
AD-




A




12145





1195-1213
1342
AUGCUCAUAGAGCAAAGA-
197
AuGcucAuAGAGcAAAGAATsT
198
UUCUUUGCUCuAUGAGcAUTsT
AD-




A




12146





1412-1430
1343
AAAAAUUGGUGCUGUUGA-
199
AAAAAuuGGuGcuGuuGAGTsT
200
CUcAAcAGcACcAAUUUUUTsT
AD-




G




12147





1431-1449
1344
GAGGAGCUGAAUAGGGUU-
201
GAGGAGcuGAAuAGGGuuATsT
202
uAACCCuAUUcAGCUCCUCTsT
AD-




A




12148





1433-1451
1345
GGAGCUGAAUAGGGUUAC-
203
GGAGcuGAAuAGGGuuAcATsT
204
UGuAACCCuAUUcAGCUCCTsT
AD-




A




12149





1434-1452
1346
GAGCUGAAUAGGGUUACA-
205
GAGcuGAAuAGGGuuAcAGTsT
206
CUGuAACCCuAUUcAGCUCTsT
AD-




G




12150





1435-1453
1347
AGCUGAAUAGGGUUACAG-
207
AGcuGAAuAGGGuuAcAGATsT
208
UCUGuAACCCuAUUcAGCUTsT
AD-




A




12151





1436-1454
1348
GCUGAAUAGGGUUACAGA-
209
GcuGAAuAGGGuuAcAGAGTsT
210
CUCUGuAACCCuAUUcAGCTsT
AD-




G




12152





1684-1702
1349
CCAAACUGGAUCGUAAGA-
211
ccAAAcuGGAucGuAAGAATsT
212
UUCUuACGAUCcAGUUUGGTsT
AD-




A




12153





1692-1710
1350
GAUCGUAAGAAGGCAGUU-
213
GAucGuAAGAAGGcAGuuGTsT
214
cAACUGCCUUCUuACGAUCTsT
AD-




G




12154





1833-1851
1351
ACCUUAUUUGGUAAUCUG-
215
AccuuAuuuGGuAAucuGcTsT
216
GcAGAUuACcAAAuAAGGUTsT
AD-




C




12155





1872-1890
1352
UUAGAUACCAUUACUACA-
217
uuAGAuAccAuuAcuAcAGTsT
218
CUGuAGuAAUGGuAUCuAATsT
AD-




G




12156





1876-1894
1353
AUACCAUUACUACAGUAG-
219
AuAccAuuAcuAcAGuAGcTsT
220
GCuACUGuAGuAAUGGuAUTsT
AD-




C




12157





1883-1901
1354
UACUACAGUAGCACUUGG-
221
uAcuAcAGuAGcAcuuGGATsT
222
UCcAAGUGCuACUGuAGuATsT
AD-




A




12158





1987-2005
1355
AAAGUAAAACUGUACUAC-
223
AAAGuAAAAcuGuAcuAcATsT
224
UGuAGuAcAGUUUuACUUUTsT
AD-




A




12159





2022-2040
1356
CUCAAGACUGAUCUUCUA-
225
cucAAGAcuGAucuucuAATsT
226
UuAGAAGAUcAGUCUUGAGTsT
AD-




A




12160





2124-2142
1357
UUGACAGUGGCCGAUAAG-
227
uuGAcAGuGGccGAuAAGATsT
228
UCUuAUCGGCcACUGUcAATsT
AD-




A




12161





2125-2143
1358
UGACAGUGGCCGAUAAGA-
229
uGAcAGuGGccGAuAAGAuTsT
230
AUCUuAUCGGCcACUGUcATsT
AD-




U




12162





2246-2264
1359
GCAAUGGGGAAACCUAAC-
231
GcAAuGuGGAAAccuAAcuTsT
232
AGUuAGGUUUCcAcAUUGCTsT
AD-




U




12163





2376-2394
1360
CCACUUAGUAGUGGCCAG-
233
ccAcuuAGuAGuGuccAGGTsT
234
CCUGGAcACuACuAAGUGGTsT
AD-




G




12164





2504-2522
1361
AGAAGGUACAAAAUUGGU-
235
AGAAGGuAcAAAAuuGGuuTsT
236
AACcAAUUUUGuACCUUCUTsT
AD-




G




12165





2852-2870
1362
UGGUUUGACUAAGCUUAA-
237
uGGuuuGAcuAAGcuuAAuTsT
238
AUuAAGCUuAGUCAAACcATsT
AD-




G




12166





2853-2871
1363
GGUUUGACUAAGCUUAAU-
239
GGuuuGAcuAAGcuuAAuuTsT
240
AAUuAAGCUuAGUcAAACCTsT
AD-




G




12167





3110-3128
1364
UCUAAGUCAAGAGCCAUC-
241
ucuAAGucAAGAGccAucuTsT
242
AGAUGGCUCUUGACUuAGATsT
AD-




U




12168





3764-3782
1365
UCAUCCCUAUAGUUCACUU
243
ucAucccuAuAGuucAcuuTsT
244
AAGUGAACuAuAGGGAUGATsT
AD-









12169





3765-3783
1366
CAUCCCUAUAGUUCACUUU
245
cAucccuAuAGuucAcuuuTsT
246
AAAGUGAACuAuAGGGAUGTsT
AD-









12170





4027-4045
1367
CCCUAGACUUCCCUAUUUC
247
cccuAGAcuucccuAuuucTsT
248
GAAAuAGGGAAGUCuAGGGTsT
AD-









12171





4031-4049
1368
AGACUUCCCUAUUUCGCUU
249
AGAcuucccuAuuucGcuuTsT
250
AAGCGAAAuAGGGAAGUCUTsT
AD-









12172





4082-4100
1369
UCACCAAACCAUUUGUAG-
251
ucAccAAAccAuuuGuAGATsT
252
UCuAcAAAUGGUUUGGUGATsT
AD-




A




12173





4272-4290
1370
UCCUUUAAGAGGCCUAAC-
253
uccuuuAAGAGGccuAAcuTsT
254
AGUuAGGCCUCUuAAAGGATsT
AD-




U




12174





4275-4293
1371
UUUAAGAGGCCUAACUCA-
255
uuuAAGAGGccuAAcucAuTsT
256
AUGAGUuAGGCCUCUuAAATsT
AD-




U




12175





4276-4294
1372
UUAAGAGGCCUAACUCAU-
257
uuAAGAGGccuAAcucAuuTsT
258
AAUGAGUuAGGCCUCUuAATsT
AD-




U




12176





4282-4300
1373
GGCCUAACUCAUUCACCCU
259
GGccuAAcucAuucAcccuTsT
260
AGGGUGAAUGAGUuAGGCCTsT
AD-









12177





4571-4589
1374
UGGUAUUUUUGAUCUGGC-
261
uGGuAuuuuuGAucuGGcATsT
262
UGCcAGAUcAAAAAuACcATsT
AD-




A




12178





4677-4695
1375
AGUUUAGUGUGUAAAGUU-
263
AGuuuAGuGuGuAAAGuuuTsT
264
AAACUUuAcAcACuAAACUTsT
AD-




U




12179





 152-170
1376
GCCAAAUUCGUCUGCGAA-
265
GccAAAuucGucuGcGAAGTsT
266
CUUCGcAGACGAAUUUGGCTsT
AD-




G




12180





 156-174
1377
AAUUCGUCUGCGAAGAAG-
267
AAuucGucuGcGAAGAAGATsT
268
UCUUCUUCGcAGACGAAUUTsT
AD-




A




12181





 491-509
1378
UGAAAGGUCACCUAAUGA-
269
uGAAAGGucAccuAAuGAATsT
270
UUcAUuAGGUGACCUUUcATsT
AD-




A




12182





 215-233
1379
CAGACCAUUUAAUUUGGC-
271
cAGAccAuuuAAuuuGGcATsT
272
UGCcAAAUuAAAUGGUCUGTsT
AD-




A




12183





 216-234
1380
AGACCAUUUAAUUUGGCA-
273
AGAccAuuuAAuuuGGcAGTsT
274
CUGCcAAAUuAAAUGGUCUTsT
AD-




G




12184





 416-434
1381
AGUUAUUAUGGGCUAUAA-
275
AGuuAuuAuGGGcuAuAAuTsT
276
AUuAuAGCCcAuAAuAACUTsT
AD-




U




12185





 537-555
1382
GCUGGUAUAAUUCCACGU-
277
GcuGGuAuAAuuccAcGuATsT
278
uACGUGGAAUuAuACcAGCTsT
AD-




A




12186





 221-239
1383
AUUUAAUUUGGCAGAGCG-
279
AuuuAAuuuGGcAGAGcGGTsT
280
CCGCUCUGCcAAAUuAAAUTsT
AD-




G




12187





 222-240
1384
UUUAAUUUGGCAGAGCGG-
281
uuuAAuuuGGcAGAGcGGATsT
282
UCCGCUCUGCcAAAUuAAATsT
AD-




A




12188





 227-245
1385
UUUGGCAGAGCGGAAAGC-
283
uuuGGcAGAGcGGAAAGcuTsT
284
AGCUUUCCGCUCUGCcAAATsT
AD-




U




12189





 476-494
1386
UUUUACAAGGGAAGGUGA-
285
uuuuAcAAGGGAAGGuGAATsT
286
UUcACCUUCcAUUGuAAAATsT
AD-




A




12190





 482-500
1387
AAUGGAAGGUGAAAGGUC-
287
AAuGGAAGGuGAAAGGucATsT
288
UGACCUUUcACCUUCcAUUTsT
AD-




A




12191





 208-226
1388
UGAGAUGCAGACCAUUUA-
289
uGAGAuGcAGAccAuuuAATsT
290
UuAAAUGGUCUGcAUCUcATsT
AD-




A




12192





 147-165
1389
UCGCAGCCAAAUUCGUCUG
291
ucGcAGccAAAuucGucuGTsT
292
cAGACGAAUUUGGCUGCGATsT
AD-









12193





 426-444
1390
GGCUAUAAUUGCACUAUC-
293
GGcuAuAAuuGcAcuAucuTsT
294
AGAuAGUGcAAUuAuAGCCTsT
AD-




U




12194





2123-2141
1391
AUUGACAGUGGCCGAUAA-
295
AuuGAcAGuGGccGAuAAGTsT
296
CUuAUCGGCcACUGUcAAUTsT
AD-




G




12195





4029-4047
1392
CUAGACUUCCCUAUUUCGC
297
cuAGAcuucccuAuuucGcTsT
298
GCGAAAuAGGGAAGUCuAGTsT
AD-









12196





 438-456
1393
ACUAUCUUUGCGUAUGGC-
299
AcuAucuuuGcGuAuGGccTsT
300
GGCcAuACGcAAAGAuAGUTsT
AD-




C




12197





 830-848
1394
AUACUCUAGUCGUUCCCAC
301
AuAcucuAGucGuucccAcTsT
302
GUGGGAACGACuAGAGuAUTsT
AD-









12198





 876-894
1395
AAAGAAACUACGAUUGAU-
303
AAAGAAAcuAcGAuuGAuGTsT
304
cAUcAAUCGuAGUUUCUUUTsT
AD-




G




12199





 115-133
1396
GCCUUGAUUUUUUGGCGG-
305
GccuuGAuuuuuuGGcGGGTsT
306
CCCGCcAAAAAAUcAAGGCTsT
AD-




G




12200





 248-266
1397
CGCCCAUUCAAUAGUAGA-
307
cGcccAuucAAuAGuAGAATsT
308
UUCuACuAUUGAAUGGGCGTsT
AD-




A




12201





1834-1852
1398
CCUUAUUUGGUAAUCUGC-
309
ccuuAuuuGGuAAucuGcuTsT
310
AGcAGAUuACcAAAuAAGGTsT
AD-




U




12202





3050-3068
1399
AGAGACAAUUCCGGAUGU-
311
AGAGAcAAuuccGGAuGuGTsT
312
cAcAUCCGGAAUUGUCUCUTsT
AD-




G




12203





4705-4723
1400
UGACUUUGAUAGCUAAAU-
313
uGAcuuuGAuAGcuAAAuuTsT
314
AAUUuAGCuAUcAAAGUcATsT
AD-




U




12204





 229-247
1401
UGGCAGAGCGGAAAGCUA-
315
uGGcAGAGcGGAAAGcuAGTsT
316
CuAGCUUUCCGCUCUGCcATsT
AD-




G




12205





 234-252
1402
GAGCGGAAAGCUAGCGCCC
317
GAGcGGAAAGcuAGcGcccTsT
318
GGGCGCuAGCUUUCCGCUCTsT
AD-









12206





 282-300
1403
AAAGAAGUUAGUGUACGA-
319
AAAGAAGuuAGuGuAcGAATsT
320
UUCGuAcACuAACUUCUUUTsT
AD-




A




12207





 433-451
1404
AUUGCACUAUCUUUGCGU-
321
AuuGcAcuAucuuuGcGuATsT
322
uACGcAAAGAuAGUGcAAUTsT
AD-




A




12208





 540-558
1405
GGUAUAAUUCCACGUACCC
323
GGuAuAAuuccAcGuAcccTsT
324
GGGuACGUGGAAUuAuACCTsT
AD-









12209





 831-849
1406
UACUCUAGUCGUUCCCACU
325
uAcucuAGucGuucccAcuTsT
326
AGUGGGAACGACuAGAGuATsT
AD-









12210





 872-890
1407
UAUGAAAGAAACUACGAU-
327
uAuGAAAGAAAcuAcGAuuTsT
328
AAUCGuAGUUUCUUUcAuATsT
AD-




U




12211





1815-1833
1408
AUGCUAGAAGUACAUAAG-
329
AuGcuAGAAGuAcAuAAGATsT
330
UCUuAUGuACUUCuAGcAUTsT
AD-




A




12212





1822-1840
1409
AAGUACAUAAGACCUUAU-
331
AAGuAcAuAAGAccuuAuuTsT
332
AAuAAGGUCUuAUGuACUUTsT
AD-




U




12213





3002-3020
1410
ACAGCCUGAGCUGUUAAU-
333
AcAGccuGAGcuGuuAAuGTsT
334
cAUuAAcAGCUcAGGCUGUTsT
AD-




G




12214





3045-3063
1411
AAAGAAGAGACAAUUCCG-
335
AAAGAAGAGAcAAuuccGGTsT
336
CCGGAAUUGUCUCUUCUUUTsT
AD-




G




12215





3224-3242
1412
CACACUGGAGAGGUCUAA-
337
cAcAcuGGAGAGGucuAAATsT
338
UUuAGACCUCUCcAGUGUGTsT
AD-




A




12216





3226-3244
1413
CACUGGAGAGGUCUAAAG-
339
cAcuGGAGAGGucuAAAGuTsT
340
ACUUuAGACCUCUCcAGUGTsT
AD-




U




12217





3227-3245
1414
ACUGGAGAGGUCUAAAGU-
341
AcuGGAGAGGucuAAAGuGTsT
342
cACUUuAGACCUCUCcAGUTsT
AD-




G




12218





 145-163
1415
CGUCGCAGCCAAAUUCGUC
343
cGucGcAGccAAAuucGucTsT
344
GACGAAUUUGGCUGCGACGTsT
AD-









12219





1700-1718
1416
GAAGGCAGUUGACCAACA-
345
GAAGGcAGuuGAccAAcAcTsT
346
GUGUUGGUcAACUGCCUUCTsT
AD-




C




12220





4291-4309
1417
CAUUCACCCUGACAGAGUU
347
cAuucAcccuGAcAGAGuuTsT
348
AACUCUGUcAGGGUGAAUGTsT
AD-









12221





4278-4296
1418
AAGAGGCCUAACUCAUUC-
349
AAGAGGccuAAcucAuucATsT
350
UGAAUGAGUuAGGCCUCUUTsT
AD-




A




12222





3051-3069
1419
GAGACAAUUCCGGAUGUG-
351
GAGAcAAuuccGGAuGuGGTsT
352
CcAcAUCCGGAAUUGUCUCTsT
AD-




G




12223





3058-3076
1420
UUCCGGAUGUGGAUGUAG-
353
uuccGGAuGuGGAuGuAGATsT
354
UCuAcAUCcAcAUCCGGAATsT
AD-




A




12224





 241-259
1421
AAGCUAGCGCCCAUUCAAU
355
AAGcuAGcGcccAuucAAuTsT
356
AUUGAAUGGGCGCuAGCUUTsT
AD-









12225





 285-303
1422
GAAGUUAGUGUACGAACU-
357
GAAGuuAGuGuAcGAAcuGTsT
358
cAGUUCGuAcACuAACUUCTsT
AD-




G




12226





 542-560
1423
UAUAAUUCCACGUACCUU
359
uAuAAuuccAcGuAccuuTsT
360
AAGGGuACGUGGAAUuAuATsT
AD-









12227





2127-2145
1424
ACAGUGGCCGAUAAGAUA-
361
AcAGuGGccGAuAAGAuAGTsT
362
CuAUCUuAUCGGCcACUGUTsT
AD-




G




12228





3760-3778
1425
UCUGUCAUCCCUAUAGUUC
363
ucuGucAucccuAuAGuucTsT
364
GAACuAuAGGGAUGAcAGATsT
AD-









12229





3993-4011
1426
UUCUUGCUAUGACUUGUG-
365
uucuuGcuAuGAcuuGuGuTsT
366
AcAcAAGUcAuAGcAAGAATsT
AD-




U




12230





1696-1714
1427
GUAAGAAGGCAGUUGACC-
367
GuAAGAAGGcAGuuGAccATsT
368
UGGUcAACUGCCUUCUuACTsT
AD-




A




12231





2122-2140
1428
CAUUGACAGUGGCCGAUA-
369
cAuuGAcAGuGGccGAuAATsT
370
UuAUCGGCcACUGUcAAUGTsT
AD-




A




12232





2371-2389
1429
AGAAACCACUUAGUAGUG-
371
AGAAAccAcuuAGuAGuGuTsT
372
AcACuACuAAGUGGUUUCUTsT
AD-




U




12233





3143-3161
1430
GGAUUGUUCAUCAAUUGG-
373
GGAuuGuucAucAAuuGGcTsT
374
GCcAAUUGAUGAAcAAUCCTsT
AD-




C




12234





4277-4295
1431
UAAGAGGCCUAACUCAUU-
375
uAAGAGGccuAAcucAuucTsT
376
GAAUGAGUuAGGCCUCUuATsT
AD-




C




12235





 287-305
1432
AGUUAGUGUACGAACUGG-
377
AGuuAGuGuAcGAAcuGGATsT
378
UCcAGUUCGuAcACuAACUTsT
AD-




A




12236





1823-1841
1433
AGUACAUAAGACCUUAUU-
379
AGuAcAuAAGAccuuAuuuTsT
380
AAAuAAGGUCUuAUGuACUTsT
AD-




U




12237





3379-3397
1434
UGAGCCUUGUGUAUAGAU-
381
uGAGccuuGuGuAuAGAuuTsT
382
AAUCuAuAcAcAAGGCUcATsT
AD-




U




12238





4273-4291
1435
CCUUUAAGAGGCCUAACUC
383
ccuuuAAGAGGccuAAcucTsT
384
GAGUuAGGCCUCUuAAAGGTsT
AD-









12239





2375-2393
1436
ACCACUUAGUAGUGUCCA-
385
AccAcuuAGuAGuGuccAGTsT
386
CUGGAcACuACuAAGUGGUTsT
AD-




G




12240





4439-4457
1437
GAAACUUCCAAUUAUGUC-
387
GAAAcuuccAAuuAuGucuTsT
388
AGAcAuAAUUGGAAGUUUCTsT
AD-




U




12241





 827-845
1438
UGCAUACUCUAGUCGUUCC
389
uGcAuAcucuAGucGuuccTsT
390
GGAACGACuAGAGuAUGcATsT
AD-









12242





1699-1717
1439
AGAAGGCAGUUGACCAAC-
391
AGAAGGcAGuuGAccAAcATsT
392
UGUUGGUcAACUGCCUUCUTsT
AD-




A




12243





1824-1842
1440
GUACAUAAGACCUUAUUU-
393
GuAcAuAAGAccuuAuuuGTsT
394
cAAAuAAGGUCUuAUGuACTsT
AD-




G




12244





 429-447
1441
UAUAAUUGCACUAUCUUU-
395
uAuAAuuGcAcuAucuuuGTsT
396
cAAAGAuAGUGcAAUuAuATsT
AD-




G




12245





 856-874
1442
UCUCUGUUACAAUACAUA-
397
ucucuGuuAcAAuAcAuAuTsT
398
AuAUGuAUUGuAAcAGAGATsT
AD-




U




12246





1194-1212
1443
UAUGCUCAUAGAGCAAAG-
399
uAuGcucAuAGAGcAAAGATsT
400
UCUUUGCUCuAUGAGcAuATsT
AD-




A




12247





 392-410
1444
UGUUGUUUGUCCAAUUCU-
401
uGuuGuuuGuccAAuucuGTsT
402
cAGAAUUGGAcAAAcAAcATsT
AD-




G




12248





1085-1103
1445
ACUAACUAGAAUCCUCCAG
403
AcuAAcuAGAAuccuccAGTsT
404
CUGGAGGAUUCuAGUuagutST
AD-









12249





2069-2087
1446
UGUGGUGUCUAUACUGAA-
405
uGuGGuGucuAuAcuGAAATsT
406
UUUcAGuAuAGAcACcAcATsT
AD-




A




12250





4341-4359
1447
UAUUAUGGGAGACCACCC-
407
uAuuAuGGGAGAccAcccATsT
408
UGGGUGGUCUCCcAuAAuATsT
AD-




A




12251





 759-777
1448
AAGGAUGAAGUCUAUCAA-
409
AAGGAuGAAGucuAucAAATsT
410
UUUGAuAGACUUcAUCCUUTsT
AD-




A




12252





 973-991
1449
UUGAUAAGAGAGCUCGGG-
411
uuGAuAAGAGAGcucGGGATsT
412
UCCCGAGCUCUCUuAUcAATsT
AD-




A




12253





1063-1081
1450
AUGUUCCUUAUCGAGAAU-
413
AuGuuccuuAucGAGAAucTsT
414
GAUUCUCGAuAAGGAAcAUTsT
AD-




C




12254





1190-1208
1451
GGAAUAUGCUCAUAGAGC-
415
GGAAuAuGcucAuAGAGcATsT
416
UGCUCuAUGAGcAuAUUCCTsT
AD-




A




12255





1679-1697
1452
CCAUUCCAAACUGGAUCGU
417
ccAuuccAAAcuGGAucGuTsT
418
ACGAUCcAGUUUGGAAUGGTsT
AD-









12256





1703-1721
1453
GGCAGUUGACCAACACAA-
419
GGcAGuuGAccAAcAcAAuTsT
420
AUUGUGUUGGUcAACUGCCTsT
AD-




U




12257





1814-1832
1454
CAUGCUAGAAGUACAUAA-
421
cAuGcuAGAAGuAcAuAAGTsT
422
CUuAUGuACUUCuAGcAUGTsT
AD-




G




12258





1818-1836
1455
CUAGAAGUACAUAAGACC-
423
cuAGAAGuAcAuAAGAccuTsT
424
AGGUCUuAUGuACUUCuAGTsT
AD-




U




12259





1897-1915
1456
UUGGAUCUCUCACAUCUA-
425
uuGGAucucucAcAucuAuTsT
426
AuAGAUGUGAGAGAUCcAATsT
AD-




U




12260





2066-2084
1457
AACUGUGGUGUCUAUACU-
427
AAcuGuGGuGucuAuAcuGTsT
428
cAGuAuAGAcACcAcAGUUTsT
AD-




G




12261





2121-2139
1458
UCAUUGACAGUGGCCGAU-
429
ucAuuGAcAGuGGccGAuATsT
430
uAUCGGCcACUGUcAAUGATsT
AD-




A




12262





2280-2298
1459
AUAAAGCAGACCCAUUCCC
431
AuAAAGcAGAcccAuucccTsT
432
GGGAAUGGGUCUGCUUuAUTsT
AD-









12263





2369-2387
1460
ACAGAAACCACUUAGUAG-
433
AcAGAAAccAcuuAGuAGGTsT
434
ACuACuAAGUGGUUUCUGUTsT
AD-




G




12264





2372-2390
1461
GAAACCACUUAGUAGUGU-
435
GAAAccAcuuAGuAGuGucTsT
436
GAcACuACuAAGUGGUUUCTsT
AD-




C




12265





2409-2427
1462
AAAUCUAAGGAUAUAGUC-
437
AAAucuAAGGAuAuAGucATsT
438
UGACuAuAUCCUuAGAUUUTsT
AD-




A




12266





2933-2951
1463
UUAUUUAUACCCAUCAAC-
439
uuAuuuAuAcccAucAAcATsT
440
UGUUGAUGGGuAuAAAuAATsT
AD-




A




12267





3211-3229
1464
ACAGAGGCAUUAACACAC-
441
AcAGAGGcAuuAAcAcAcuTsT
442
AGUGUGUuAAUGCCUCUGUTsT
AD-




U




12268





3223-3241
1465
ACACACUGGAGAGGUCUA-
443
AcAcAcuGGAGAGGucuAATsT
444
UuAGACCUCUCcAGUGUGUTsT
AD-




A




12269





3225-3243
1466
ACACUGGAGAGGUCUAAA-
445
AcAcuGGAGAGGucuAAAGTsT
446
CUUuAGACCUCUCcAGUGUTsT
AD-




G




12270





3291-3309
1467
CGAGCCCAGAUCAACCUUU
447
cGAGcccAGAucAAccuuuTsT
448
AAAGGUUGAUCUGGGCUCGTsT
AD-









12271





4036-4054
1468
UCCCUAUUUCGCUUUCUCC
449
ucccuAuuucGcuuucuccTsT
450
GGAGAAAGCGAAAuAGGGATsT
AD-









12272





4180-4198
1469
UCUAAAAUCACUGUCAAC-
451
ucuAAAAucAcuGucAAcATsT
452
UGUUGAcAGUGAUUUuAGATsT
AD-




A




12273





 151-169
1470
AGCCAAAUUCGUCUGCGA-
453
AGccAAAuucGucuGcGAATsT
454
UUCGcAGACGAAUUUGGCUTsT
AD-




A




12274





 250-268
1471
CCCAUUCAAUAGUAGAAU-
455
cccAuucAAuAGuAGAAuGTsT
456
cAUUCuACuAUUGAAUGGGTsT
AD-




G




12275





 821-839
1472
GAUGAAUGCAUACUCUAG-
457
GAuGAAuGcAuAcucuAGuTsT
458
ACuAGAGuAUGcAUUcAUCTsT
AD-




U




12276





1060-1078
1473
CUCAUGUUCCUUAUCGAG-
459
cucAuGuuccuuAucGAGATsT
460
UCUCGAuAAGGAAcAUGAGTsT
AD-




A




12277





1075-1093
1474
GAGAAUCUAAACUAACUA-
461
GAGAAucuAAAcuAAcuAGTsT
462
CuAGUuAGUUuAGAUUCUCTsT
AD-




G




12278





1819-1837
1475
UAGAAGUACAUAAGACCU-
463
uAGAAGuAcAuAAGAccuuTsT
464
AAGGUCUuAUGuACUUCuATsT
AD-




U




12279





3003-3021
1476
CAGCCUGAGCUGUUAAUG-
465
cAGccuGAGcuGuuAAuGATsT
466
UcAUuAAcAGCUcAGGCUGTsT
AD-




A




12280





3046-3064
1477
AAGAAGAGACAAUUCCGG-
467
AAGAAGAGAcAAuuccGGATsT
468
UCCGGAAUUGUCUCUUCUUTsT
AD-




A




12281





3134-3152
1478
UGCUGGUGUGGAUUGUUC-
469
uGcuGGuGuGGAuuGuucATsT
470
UGAAcAAUCcAcACcAGcATsT
AD-




A




12282





 155-173
1479
AAAUUCGUCUGCGAAGAA-
471
AAAuucGucuGcGAAGAAGTsT
472
CUUCUUCGcAGACGAAUUUTsT
AD-




G




12283





4596-4614
1480
UUUCUGGAAGUUGAGAUG-
473
uuucuGGAAGuuGAGAuGuTsT
474
AcAUCUcAACUUCcAGAAATsT
AD-




U




12284





 365-383
1481
UACUAAACAGAUUGAUGU-
475
uAcuAAAcAGAuuGAuGuuTsT
476
AAcAUcAAUCUGUUuAGuATsT
AD-




U




12285





 374-392
1482
GAUUGAUGUUUACCGAAG-
477
GAuuGAuGuuuAccGAAGuTsT
478
ACUUCGGuAAAcAUcAAUCTsT
AD-




U




12286





 436-454
1483
GCACUAUCUUUGCGUAUG-
479
GcAcuAucuuuGcGuAuGGTsT
480
CcAuACGcAAAGAuAGUGCTsT
AD-




G




12287





 539-557
1484
UGGUAUAAUUCCACGUAC-
481
uGGuAuAAuuccAcGuAccTsT
482
GGuACGUGGAAUuAuACcATsT
AD-




C




12288





1629-1647
1485
AGCAAGCUGCUUAACACA-
483
AGcAAGcuGcuuAAcAcAGTsT
484
CUGUGUuAAGcAGCUUGCUTsT
AD-




G




12289





2370-2388
1486
CAGAAACCACUUAGUAGU-
485
cAGAAAccAcuuAGuAGuGTsT
486
cACuACuAAGUGGUUUCUGTsT
AD-




G




12290





2676-2694
1487
AACUUAUUGGAGGUUGGA-
487
AAcuuAuuGGAGGuuGGAATsT
488
UuAcAACCUCcAAuAAGUUTsT
AD-




A




12291





3228-3246
1488
CUGGAGAGGUCUAAAGUG-
489
cuGGAGAGGucuAAAGuGGTsT
490
CcACUUuAGACCUCUCcAGTsT
AD-




G




12292





3703-3721
1489
AAAAAAGAUAUAAGGCAG-
491
AAAAAAGAuAuAAGGcAGuTsT
492
ACUGCCUuAuAUCUUUUUUTsT
AD-




U




12293





3737-3755
1490
GAAUUUUGAUAUCUACCC-
493
GAAuuuuGAuAucuAcccATsT
494
UGGGuAGAuAUcAAAAUUCTsT
AD-




A




12294





4573-4591
1491
GUAUUUUUGAUCUGGCAA-
495
GuAuuuuuGAucuGGcAAcTsT
496
GUUGCcAGAUcAAAAAuACTsT
AD-




C




12295





 526-544
1492
AGGAUCCCUUGGCUGGUA-
497
AGGAucccuuGGcuGGuAuTsT
498
AcACcAGCcAAGGGAUCCUTsT
AD-




U




12296





 527-545
1493
GGAUCCCUUGGCUGGUAU-
499
GGAucccuuGGcuGGuAuATsT
500
uAuACcAGCcAAGGGAUCCTsT
AD-




A




12297





 256-274
1494
CAAUAGUAGAAUGUGAUC-
501
cAAuAGuAGAAuGuGAuccTsT
502
GGAUcAcAUUCrACuAUUGTsT
AD-




C




12298





 427-445
1495
GCUAUAAUUGCACUAUCU-
503
GcuAuAAuuGcAcuAucuuTsT
504
AAGAuAGUGcAAUuAuAGCTsT
AD-




U




12299





 554-572
1496
UACCCUUCAUCAAAUUUU-
505
uAcccuucAucAAAuuuuuTsT
506
AAAAAUUUGAUGAAGGGuATsT
AD-




U




12300





1210-1228
1497
AGAACAUAUUGAAUAAGC-
507
AGAAcAuAuuGAAuAAGccTsT
508
GGCUuAUUcAAuAUGUUCUTsT
AD-




C




12301





1414-1432
1498
AAAUUGGUGCUGUUGAGG-
509
AAAuuGGuGcuGuuGAGGATsT
510
UCCUcAAcAGcACcAAUUUTsT
AD-




A




12302





1438-1456
1499
UGAAUAGGGUUACAGAGU-
511
uGAAuAGGGuuAcAGAGuuTsT
512
AACUCUGuAACCCuAUUcATsT
AD-




U




12303





1516-1534
1500
AAGAACUUGAAACCACUC-
513
AAGAAcuuGAAAccAcucATsT
514
UGAGUGGUUUcAAGUUCUUTsT
AD-




A




12304





2279-2297
1501
AAUAAAGCAGACCCAUUCC
515
AAuAAAGcAGAcccAuuccTsT
516
GGAAUGGGUCUGCUUuAUUTsT
AD-









12305





2939-2957
1502
AUACCCAUCAACACUGGUA
517
AuAcccAucAAcAcuGGuATsT
518
uACcAGUGUUGAUGGGuAUTsT
AD-









12306





3142-3160
1503
UGGAUUGUUCAUCAAUUG-
519
uGGAuuGuucAucAAuuGGTsT
520
CcAAUUGAUGAAcAAUCcATsT
AD-




G




12307





3229-3247
1504
UGGAGAGGUCUAAAGUGG-
521
uGGAGAGGucuAAAGuGGATsT
522
UCcACUUuAGACCUCUCcATsT
AD-




A




12308





3763-3781
1505
GUCUACCCUAUAGUUCACU
523
GucuAcccuAuAGuucAcuTsT
524
AGUGAACuAuAGGGAUGACTsT
AD-









12309





4801-4819
1506
AUAAUGGCUAUAAUUUCU-
525
AuAAuGGcuAuAAuuucucTsT
526
GAGAAAUuAuAGCcAUuAUTsT
AD-




C




12310





 529-547
1507
AUCCCUUGGCUGGUAUAA-
527
AucccuuGGcuGGuAuAAuTsT
528
AUuAuACuAGCcAAGGGAUTsT
AD-




U




12311





 425-443
1508
GCGCUAUAAUUGCACUAU-
529
GcGcuAuAAuuGcAcuAucTsT
530
GAuAGUGcAAUuAuAGCCCTsT
AD-




C




12312





1104-1122
1509
GAUUCUCUGGGAGGGCGU-
531
GAuucucuGGGAGGGcGuATsT
532
uACGCCCUCcAAGAGAAUCTsT
AD-




A




12313





1155-1173
1510
GCAUCUCUCAAUCUUGAG-
533
GcAucucucAAucuuGAGGTsT
534
CCUcAAGAUUGAGAGAUGCTsT
AD-




G




12314





2403-2421
1511
CAGCAGAAAUCUAAGGAU-
535
cAGcAGAAAucuAAGGAuATsT
536
uAUCCUuAGAUUUCUGCUGTsT
AD-




A




12315





3115-3133
1512
GUCAAGAGCCAUCUGUAG-
537
GucAAGAGccAucuGuAGATsT
538
UCuAcAGAUGGCUCUUGACTsT
AD-




A




12316





3209-3227
1513
AAACAGAGGCAUUAACAC-
539
AAAcAGAGGcAuuAAcAcATsT
540
UGUGUuAAUGCCUCUGUUUTsT
AD-




A




12317





3293-3311
1514
AGCCCAGAUCAACCUUUAA
541
AGcccAGAucAAccuuuAATsT
542
UuAAAGGUUGAUCUGGGCUTsT
AD-









12318





4574-4592
1515
UAUUUUUGAUCUGGCAAC-
543
uAuuuuuGAucuGGcAAccTsT
544
GGUUGCcAGAUcAAAAAuATsT
AD-




C




12319





 352-370
1516
UGUGUGGAGCAUCUACUA-
545
uGuGuGGAGcAucuAcuAATsT
546
UuAGuAGAUGCUCcAAAcATsT
AD-




A




12320





 741-759
1517
GAAAUUACAGUACACAAC-
547
GAAAuuAcAGuAcAcAAcATsT
548
UGUUGUGuACUGuAAUUUCTsT
AD-




A




12321





1478-1496
1518
ACUUGACCAGUGUAAAUC-
549
AcuuGAccAGuGuAAAucuTsT
550
AGAUUuAcACUGGUcAAGUTsT
AD-




U




12322





1483-1501
1519
ACCAGUGUAAAUCUGACC-
551
AccAGuGuAAAucuGAccuTsT
552
AGGUcAGAUUuAcACUGGUTsT
AD-




U




12323





1967-1985
1520
AGAACAAUCAUUAGCAGC-
553
AGAAcAAucAuuAGcAGcATsT
554
UGCUGCuAAUGAUUGUUCUTsT
AD-




A




12324





2247-2265
1521
CAAUGUGGAAACCUAACU-
555
cAAuGuGGAAAccuAAcuGTsT
556
cAGUuAGGUUUCcAcAUUGTsT
AD-




G




12325





2500-2518
1522
ACCAAGAAGGUACAAAAU-
557
AccAAGAAGGuAcAAAAuuTsT
558
AAUUUUGuACCUUCUUGGUTsT
AD-




U




12326





2508-2526
1523
GGUACAAAAUUGGUUGAA-
559
GGuAcAAAAuuGGuuGAAGTsT
560
CUUcAACcAAUUUUGuACCTsT
AD-




G




12327





3138-3156
1524
GGUGUGGAUUGUUCAUCA-
561
GGuGuGGAuuGuucAucAATsT
562
UUGAUGAAcAAUCcAcACCTsT
AD-




A




12328





4304-4322
1525
AGAGUUCACAAAAAGCCC-
563
AGAGuucAcAAAAAGcccATsT
564
UGGGCUUUUUGUGAACUCUTsT
AD-




A




12329





4711-4729
1526
UGAUAGCUAAAUUAAACC-
565
uGAuAGcuAAAuuAAAccATsT
566
UGGUUuAAUUuAGCuAUcATsT
AD-




A




12330





1221-1239
1527
AAUAAGCCUGAAGUGAAU-
567
AAuAAGccuGAAGuGAAucTsT
568
GAUUcACUUcAGGCUuAUUTsT
AD-




C




12331





1705-1723
1528
CAGUUGACCAACACAAUGC
569
cAGuuGAccAAcAcAAuGcTsT
570
GcAUUGUGUUGGUcAACUGTsT
AD-









12332





3137-3155
1529
UGGUGUGGAUUGUUCAUC-
571
uGGuGuGGAuuGuucAucATsT
572
UGAUGAAcAAUCcAcACcATsT
AD-




A




12333





4292-4310
1530
AUUCACCCUGACAGAGUUC
573
AuucAcccuGAcAGAGuucTsT
574
GAACUCUGUcAGGGUGAAUTsT
AD-









12334





1829-1847
1531
UAAGACCUUAUUUGGUAA-
575
uAAGAccuuAuuuGGuAAuTsT
576
AUuACcAAAuAAGGUCUuATsT
AD-




U




12335





2244-2262
1532
AAGCAAUGUGGAAACCUA-
577
AAGcAAuGuGGAAAccuAATsT
578
UuAGGUUUCcAcAUUGCUUTsT
AD-




A




12336





2888-2906
1533
UCUGAAACUGGAUAUCCC-
579
ucuGAAAcuGGAuAucccATsT
580
UGGGAuAUCcAGUUUcAGATsT
AD-




A


























TABLE 2B






1st

2nd






single

single



dose

dose

3rd
SDs 3rd



screen @
SDs 1st
screen @
SDs 2nd
single
screen



50 nM [%
screen
25 nM [%
screen
dose
(among


duplex
resudual
(among
resudual
(among
screen
quadrup-


name
mRNA]
quadruplicates)
mRNA]
quadruplicates)
@25 nM
licates)





















AD-12072
65%
2%
82%
5%




AD-12073
84%
1%
61%
6%


AD-12074
51%
3%
36%
9%


AD-12075
56%
4%
36%
4%


AD-12076
21%
4%
13%
3%


AD-12077
11%
2%
6%
1%


AD-12078
22%
3%
9%
2%


AD-12079
22%
10%
15%
7%


AD-12080
68%
4%
52%
13%


AD-12081
34%
8%
35%
24%


AD-12082
20%
2%
92%
5%


AD-12083
85%
6%
63%
10%


AD-12084
18%
6%
17%
4%


AD-12085
13%
4%
12%
4%


AD-12086
26%
5%
17%
3%


AD-12087
95%
4%
80%
4%


AD-12088
29%
6%
29%
2%


AD-12089
69%
5%
64%
7%


AD-12090
46%
15%
34%
5%


AD-12091
16%
6%
17%
3%


AD-12092
82%
26%
63%
5%


AD-12093
84%
4%
70%
4%


AD-12094
46%
3%
34%
3%


AD-12095
14%
2%
13%
1%


AD-12096
26%
11%
17%
1%


AD-12097
23%
2%
21%
1%


AD-12098
41%
14%
17%
3%


AD-12099
57%
2%
48%
6%


AD-12100
101%
11%
98%
8%


AD-12101
46%
7%
32%
2%


AD-12102
96%
17%
88%
18%


AD-12103
19%
5%
20%
2%


AD-12104
40%
8%
24%
2%


AD-12105
39%
2%
35%
10%


AD-12106
87%
6%
79%
19%


AD-12107
29%
2%
32%
16%


AD-12108
38%
4%
39%
8%


AD-12109
49%
3%
44%
10%


AD-12110
85%
5%
80%
14%


AD-12111
64%
6%
71%
18%


AD-12112
48%
4%
41%
5%


AD-12113
13%
0%
14%
3%


AD-12114
32%
6%
16%
4%


AD-12115
8%
4%
7%
5%


AD-12116
74%
5%
61%
7%


AD-12117
21%
4%
20%
2%


AD-12118
44%
4%
42%
6%


AD-12119
37%
4%
24%
3%


AD-12120
22%
2%
15%
4%


AD-12121
32%
1%
22%
2%


AD-12122
36%
16%
10%
5%


AD-12123
28%
1%
16%


AD-12124
28%
2%
16%


AD-12125
15%
1%
14%


AD-12126
51%
22%
27%


AD-12127
54%
4%
42%
9%


AD-12128
29%
1%
20%
2%


AD-12129
22%
3%
19%
3%


AD-12130
53%
6%
42%
7%


AD-12131
28%
5%
22%
3%


AD-12132
88%
2%
90%
18%


AD-12133
34%
2%
26%
6%


AD-12134
18%
3%
14%
2%


AD-12135
50%
6%
37%
4%


AD-12136
42%
19%
22%
2%


AD-12137
85%
12%
92%
4%


AD-12138
47%
6%
49%
1%


AD-12139
80%
5%
72%
4%


AD-12140
97%
22%
67%
9%


AD-12141
120%
4%
107%
10%


AD-12142
55%
8%
33%
4%


AD-12143
64%
34%
19%
2%


AD-12144
58%
29%
17%
2%


AD-12145
27%
8%
18%
2%


AD-12146
19%
20%
15%
1%


AD-12147
29%
9%
35%
3%


AD-12148
30%
3%
56%
5%


AD-12149
8%
2%
12%
3%


AD-12150
31%
2%
31%
7%


AD-12151
9%
5%
14%
2%


AD-12152
3%
3%
23%
3%


AD-12153
20%
6%
34%
4%


AD-12154
24%
7%
44%
3%


AD-12155
33%
6%
53%
11%


AD-12156
35%
5%
40%
5%


AD-12157
8%
3%
23%
4%


AD-12158
13%
2%
22%
5%


AD-12159
34%
6%
46%
5%


AD-12160
19%
3%
31%
4%


AD-12161
88%
4%
83%
7%


AD-12162
26%
7%
32%
7%


AD-12163
55%
9%
40%
3%


AD-12164


21%
3%


AD-12165
30%
3%
41%
4%


AD-12166
9%
10%
22%
9%


AD-12167
26%
3%
30%
2%


AD-12168
54%
4%
59%
20%


AD-12169
41%
4%
51%
16%


AD-12170
43%
4%
52%
20%


AD-12171
67%
3%
73%
25%


AD-12172
53%
15%
37%
2%


AD-12173
39%
0%
39%
0%


AD-12174
41%
5%
27%
0%


AD-12175
29%
0%
38%
14%


AD-12176
43%
2%
56%
25%


AD-12177
68%
6%
74%
30%


AD-12178
41%
4%
41%
6%


AD-12179
53%
5%
44%
5%


AD-12180
16%
2%
13%
4%


AD-12181
19%
3%
14%
2%


AD-12182
16%
4%
18%
8%


AD-12183
26%
3%
19%
4%


AD-12184
54%
2%
77%
8%


AD-12185
8%
1%
9%
1%


AD-12186
35%
3%
41%
6%


AD-12187
34%
17%
27%
1%


AD-12188
30%
3%
27%
4%


AD-12189
51%
4%
48%
5%


AD-12190
33%
2%
26%
4%


AD-12191
20%
2%
13%
0%


AD-12192
21%
1%
23%
10%


AD-12193
64%
8%
98%
6%


AD-12194
8%
2%
15%
4%


AD-12195
34%
2%
48%
3%


AD-12196
34%
2%
51%
3%


AD-12197
75%
4%
93%
6%


AD-12198
55%
5%
48%
2%


AD-12199
102%
6%
118%
9%


AD-12200
75%
6%
60%
12%


AD-12201
42%
3%
16%
4%


AD-12202
29%
4%
9%
3%


AD-12203
114%
14%
89%
20%


AD-12204
64%
7%
26%
5%


AD-12205
66%
12%
35%
4%


AD-12206
46%
3%
32%
12%


AD-12207
57%
5%
40%
6%


AD-12208
30%
8%
10%
5%


AD-12209
101%
6%
102%
23%


AD-12210
38%
11%
27%
14%


AD-12211
16%
6%
10%
5%


AD-12212
59%
8%
65%
5%


AD-12213
24%
9%
12%
2%


AD-12214
67%
14%
70%
12%


AD-12215
29%
13%
13%
4%


AD-12216
36%
4%
13%
1%


AD-12217
36%
9%
11%
2%


AD-12218
35%
5%
17%
3%


AD-12219
41%
9%
14%
1%


AD-12220
37%
5%
23%
3%


AD-12221
58%
7%
39%
6%


AD-12222
74%
9%
53%
3%


AD-12223
74%
10%
67%
7%


AD-12224
24%
2%
11%
2%


AD-12225
75%
5%
76%
14%


AD-12226
45%
8%
40%
3%


AD-12227
61%
6%
47%
5%


AD-12228
28%
3%
25%
5%


AD-12229
54%
13%
37%
6%


AD-12230
70%
17%
65%
4%


AD-12231
32%
12%
22%
6%


AD-12232
30%
3%
17%
2%


AD-12233
38%
2%
32%
3%


AD-12234
90%
5%
95%
7%


AD-12235
57%
7%
46%
3%


AD-12236
34%
8%
16%
2%


AD-12237
42%
9%
32%
8%


AD-12238
42%
6%
34%
6%


AD-12239
42%
3%
40%
4%


AD-12240
47%
6%
36%
5%


AD-12241
69%
5%
70%
8%


AD-12242
61%
2%
47%
3%


AD-12243
26%
7%
15%
1%


AD-12244
25%
6%
15%
1%


AD-12245
65%
6%
83%
13%


AD-12246
29%
7%
31%
6%


AD-12247
57%
13%
50%
3%


AD-12248
36%
8%
20%
3%
15%
7%


AD-12249
44%
3%
70%
11%
103%
34%


AD-12250
47%
5%
18%
5%
17%
4%


AD-12251
121%
28%
35%
8%
60%
42%


AD-12252
94%
15%
8%
3%
5%
3%


AD-12253
94%
33%
42%
8%
49%
27%


AD-12254
101%
58%
70%
5%
80%
32%


AD-12255
163%
37%
38%
6%
36%
10%


AD-12256
112%
62%
18%
3%
9%
4%


AD-12257
10%
4%
9%
2%
6%
2%


AD-12258
27%
9%
18%
3%
20%
6%


AD-12259
20%
5%
12%
2%
13%
5%


AD-12260
22%
7%
81%
7%
65%
13%


AD-12261
122%
11%
66%
7%
80%
22%


AD-12262
57%
30%
33%
6%
44%
18%


AD-12263
177%
57%
85%
11%
84%
15%


AD-12264
37%
6%
10%
1%
10%
4%


AD-12265
40%
8%
17%
1%
20%
10%


AD-12266
33%
9%
9%
1%
8%
4%


AD-12267
34%
13%
11%
1%
6%
2%


AD-12268
34%
6%
11%
1%
9%
2%


AD-12269
54%
6%
33%
4%
29%
7%


AD-12270
52%
5%
29%
4%
27%
6%


AD-12271
53%
7%
27%
3%
19%
6%


AD-12272
85%
15%
57%
7%
51%
16%


AD-12273
36%
6%
26%
2%
30%
5%


AD-12274
75%
21%
40%
2%
50%
19%


AD-12275
29%
9%
8%
1%
8%
4%


AD-12276
45%
19%
15%
2%
16%
12%


AD-12277
58%
17%
32%
2%
55%
14%


AD-12278
120%
35%
96%
10%
124%
38%


AD-12279
47%
29%
17%
1%
12%
4%


AD-12280
2%
0%
3%
1%


AD-12281
2%
0%
5%
2%


AD-12282
3%
0%
25%
5%


AD-12283
3%
1%
35%
4%


AD-12284
5%
2%
49%
8%


AD-12285
7%
7%
21%
26%


AD-12286
28%
34%
12%
7%


AD-12287
40%
21%
51%
23%


AD-12288
26%
7%
155%
146%


AD-12289
43%
21%
220%
131%


AD-12290
2%
1%
81%
23%


AD-12291
4%
1%
70%
3%


AD-12292
2%
3%
6%
2%


AD-12293
4%
2%
36%
3%


AD-12294
10%
6%
38%
3%


AD-12295
29%
31%
37%
3%


AD-12296
82%
4%
89%
2%


AD-12297
75%
3%
65%
2%


AD-12298
73
4%
60%
3%


AD-12299
76%
4%
65%
4%


AD-12300
36%
4%
15%
1%


AD-12301
33%
4%
18%
2%


AD-12302
66%
5%
65%
3%


AD-12303
35%
6%
17%
2%


AD-12304
70%
8%
70%
6%


AD-12305
63%
8%
80%
7%


AD-12306
23%
6%
20%
3%


AD-12307
78%
10%
58%
5%


AD-12308
27%
8%
15%
2%


AD-12309
58%
11%
42%
3%


AD-12310
106%
23%
80%
2%


AD-12311
73%
12%
60%
2%


AD-12312
39%
3%
36%
3%


AD-12313
64%
9%
49%
6%


AD-12314
28%
7%
14%
6%


AD-12315
31%
7%
13%
2%


AD-12316
42%
5%
14%
2%


AD-12317
34%
9%
15%
5%


AD-12318
46%
4%
28%
4%


AD-12319
77%
3%
56%
4%


AD-12320
55%
7%
41%
3%


AD-12321
21%
3%
10%
2%


AD-12322
27%
8%
30%
12%


AD-12323
26%
7%
35%
18%


AD-12324
27%
8%
27%
14%


AD-12325
32%
12%
32%
22%


AD-12326
42%
22%
45%
41%


AD-12327
36%
14%
37%
32%


AD-12328
45%
2%
31%
3%


AD-12329
61%
4%
34%
3%


AD-12330
63%
5%
38%
4%


AD-12331
50%
2%
26%
5%


AD-12332
80%
4%
51%
7%


AD-12333
34%
6%
12%
2%


AD-12334
27%
2%
18%
3%


AD-12335
84%
6%
60%
7%


AD-12336
45%
4%
36%
4%


AD-12337
30%
7%
19%
2%






















TABLE 3










single
SDs







dose
2nd







screen @
screen



SEQ

SEQ

25 nM [%
(among



ID

ID
duplex
residual
quadru-


sequence (5′-3′)
NO.
sequence (5′-3′)
NO.
name
mRNA]
plicates)





















ccAuuAcuAcAGuAGcAcuTsT
582
AGUGCuACUGuAGuAAUGGTsT
583
AD-14085
19%
1%


AucuGGcAAccAuAuuucuTsT
584
AGAAAuAUGGUUGCcAGAUTsT
585
AD-14086
38%
1%


GAuAGcuAAAuuAAAccAATsT
586
UUGGUUuAAUUuAGCuAUCTsT
587
AD-14087
75%
10% 


AGAuAccAuuAcuAcAGuATsT
588
uACUGuAGuAAUGGuAUCUTsT
589
AD-14088
22%
8%


GAuuGuucAucAAuuGGcGTsT
590
CGCcAAUUGAUGAAcAAUCTsT
591
AD-14089
70%
12% 


GcuuucuccucGGcucAcuTsT
592
AGuGAGCCGAGGAGAAAGCTsT
593
AD-14090
79%
11% 


GGAGGAuuGGcuGAcAAGATsT
594
UCUUGUcAGCcAAUCCUCCTsT
595
AD-14091
29%
3%


uAAuGAAGAGuAuAccuGGTsT
596
CcAGGuAuACUCUUcAUuATsT
597
AD-14092
23%
2%


uuucAccAAAccAuuuGuATsT
598
uAcAAAUGGUUUGGUGAAATsT
599
AD-14093
60%
2%


cuuAuuAAGGAGuAuAcGGTsT
600
CCGuAuACUCCUuAAuAAGTsT
601
AD-14094
11%
3%


GAAAucAGAuGGAcGuAAGTsT
602
CUuACGUCcAUCUGAUUUCTsT
603
AD-14095
10%
2%


cAGAuGucAGcAuAAGcGATsT
604
UCGCUuAUGCUGAcAUCUGTsT
605
AD-14096
27%
2%


AucuAAcccuAGuuGuAucTsT
606
GAuAcAACuAGGGUuAGAUTsT
607
AD-14097
45%
6%


AAGAGcuuGuuAAAAucGGTsT
608
CCGAUUUuAAcAAGCUCUUTsT
609
AD-14098
50%
10% 


uuAAGGAGuAuAcGGAGGATsT
610
UCCUCCGuAuACUCCUuAATsT
611
AD-14099
12%
4%


uuGcAAuGuAAAuAcGuAuTsT
612
AuACGuAUUuAcAUUGcAATsT
613
AD-14100
49%
7%


ucuAAcccuAGuuGuAuccTsT
614
GGAuAcAACuAGGGUuAGATsT
615
AD-14101
36%
1%


cAuGuAucuuuuucucGAuTsT
616
AUCGAGAAAAAGAuAcAUGTsT
617
AD-14102
49%
3%


GAuGucAGcAuAAGcGAuGTsT
618
cAUCGCUuAUGCUGAcAUCTsT
619
AD-14103
74%
5%


ucccAAcAGGuAcGAcAccTsT
620
GGUGUCGuACCUGUUGGGATsT
621
AD-14104
27%
3%


uGcucAcGAuGAGuuuAGuTsT
622
ACuAAACUcAUCGUGAGcATsT
623
AD-14105
34%
4%


AGAGcuuGuuAAAAucGGATsT
624
UCCGAUUUuAAcAAGCUCUTsT
625
AD-14106
 9%
2%


GcGuAcAAGAAcAucuAuATsT
626
uAuAGAUGUUCUUGuACGCTsT
627
AD-14107
 5%
1%


GAGGuuGuAAGccAAuGuuTsT
628
AAcAUUGGCUuAcAACCUCTsT
629
AD-14108
15%
1%


AAcAGGuAcGAcAccAcAGTsT
630
CUGUGGUGUCGuACCUGUUTsT
631
AD-14109
91%
2%


AAcccuAGuuGuAucccucTsT
632
GAGGGAuAcAACuAGGGUUTsT
633
AD-14110
66%
5%


GcAuAAGcGAuGGAuAAuATsT
634
uAUuAUCcAUCGCUuAUGCTsT
635
AD-14111
33%
3%


AAGcGAuGGAuAAuAccuATsT
636
uAGGuAUuAUCcAUCGCUUTsT
637
AD-14112
51%
3%


uGAuccuGuAcGAAAAGAATsT
638
UUCUUUUCGuAcAGGAUcATsT
639
AD-14113
22%
3%


AAAAcAuuGGccGuucuGGTsT
640
CcAGAACGGCcAAUGUUUUTsT
641
AD-14114
117% 
8%


cuuGGAGGGcGuAcAAGAATsT
642
UUCUUGuACGCCCUCcAAGTsT
643
AD-14115
50%
8%


GGcGuAcAAGAAcAucuAuTsT
644
AuAGAUGUUCUUGuACGCCTsT
645
AD-14116
14%
3%


AcucuGAGuAcAuuGGAAuTsT
646
AUUCcAAUGuACUcAGAGUTsT
647
AD-14117
12%
4%


uuAuuAAGGAGuAuAcGGATsT
648
UCCGuAuACUCCUuAAuAATsT
649
AD-14118
26%
4%


uAAGGAGuAuAcGGAGGAGTsT
650
CUCCUCCGuAuACUCCUuATsT
651
AD-14119
24%
5%


AAAucAAuAGucAAcuAAATsT
652
UUuAGUUGACuAUUGAUUUTsT
653
AD-14120
 8%
1%


AAucAAuAGucAAcuAAAGTsT
654
CUUuAGUUGACuAUUGAUUTsT
655
AD-14121
24%
2%


uucucAGuAuAcuGuGuAATsT
656
UuAcAcAGuAuACUGAGAATsT
657
AD-14122
10%
1%


uGuGAAAcAcucuGAuAAATsT
658
UUuAUcAGAGUGUUUcAcATsT
659
AD-14123
 8%
1%


AGAuGuGAAucucuGAAcATsT
660
UGUUcAGAGAUUcAcAUCUTsT
661
AD-14124
 9%
2%


AGGuuGuAAGccAAuGuuGTsT
662
cAAcAUUGGCUuAcAACCUTsT
663
AD-14125
114% 
6%


uGAGAAAucAGAuGGAcGuTsT
664
ACGUCcAUCUGAUUUCUcATsT
665
AD-14126
 9%
1%


AGAAAucAGAuGGAcGuAATsT
666
UuACGUCcAUCUGAUUUCUTsT
667
AD-14127
57%
6%


AuAucccAAcAGGuAcGAcTsT
668
GUCGuACCUGUUGGGAuAUTsT
669
AD-14128
104% 
6%


cccAAcAGGuAcGAcAccATsT
670
UGGUGUCGuACCUGUUGGGTsT
671
AD-14129
21%
2%


AGuAuAcuGAAGAAccucuTsT
672
AGAGGUUCUUcAGuAuACUTsT
673
AD-14130
57%
6%


AuAuAuAucAGccGGGcGcTsT
674
GCGCCCGGCUGAuAuAuAUTsT
675
AD-14131
93%
6%


AAucuAAcccuAGuuGuAuTsT
676
AuAcAACuAGGGUuAGAUUTsT
677
AD-14132
75%
8%


cuAAcccuAGuuGuAucccTsT
678
GGGAuAcAACuAGGGUuAGTsT
679
AD-14133
66%
4%


cuAGuuGuAucccuccuuuTsT
680
AAAGGAGGGAuAcAACuAGTsT
681
AD-14134
44%
6%


AGAcAucuGAcuAAuGGcuTsT
682
AGCcAUuAGUcAGAUGUCUTsT
683
AD-14135
55%
6%


GAAGcucAcAAuGAuuuAATsT
684
UuAAAUcAUUGUGAGCUUCTsT
685
AD-14136
29%
3%


AcAuGuAucuuuuucucGATsT
686
UCGAGAAAAAGAuAcAUGUTsT
687
AD-14137
40%
3%


ucGAuucAAAucuuAAcccTsT
688
GGGUuAAGAUUUGAAUCGATsT
689
AD-14138
39%
5%


ucuuAAcccuuAGGAcucuTsT
690
AGAGUCCuAAGGGUuAAGATsT
691
AD-14139
71%
11% 


GcucAcGAuGAGuuuAGuGTsT
692
cACuAAACUcAUCGUGAGCTsT
693
AD-14140
43%
15% 


cAuAAGcGAuGGAuAAuAcTsT
694
GuAUuAUCcAUCGCUuAUGTsT
695
AD-14141
33%
6%


AuAAGcGAuGGAuAAuAccTsT
696
GGuAUuAUCcAUCGCUuAUTsT
697
AD-14142
51%
14% 


ccuAAuAAAcuGcccucAGTsT
698
CUGAGGGcAGUUuAUuAGGTsT
699
AD-14143
42%
1%


ucGGAAAGuuGAAcuuGGuTsT
700
ACcAAGUUcAACUUUCCGATsT
701
AD-14144
 4%
4%


GAAAAcAuuGGccGuucuGTsT
702
cAGAACGGCcAAUGUUUUCTsT
703
AD-14145
92%
5%


AAGAcuGAucuucuAAGuuTsT
704
AACUuAGAAGAUcAGUCUUTsT
705
AD-14146
13%
2%


GAGcuuGuuAAAAucGGAATsT
706
UUCCGAUUUuAAcAAGCUCTsT
707
AD-14147
 8%
1%


AcAuuGGccGuucuGGAGcTsT
708
GCUCcAGAACGGCcAAUGUTsT
709
AD-14148
80%
7%


AAGAAcAucuAuAAuuGcATsT
710
UGcAAUuAuAGAUGUUCUUTsT
711
AD-14149
44%
7%


AAAuGuGucuAcucAuGuuTsT
712
AAcAUGAGuAGAcAcAUUUTsT
713
AD-14150
32%
29% 


uGucuAcucAuGuuucucATsT
714
UGAGAAAcAUGAGuAGAcATsT
715
AD-14151
75%
11% 


GuAuAcuGuGuAAcAAucuTsT
716
AGAUUGUuAcAcAGuAuACTsT
717
AD-14152
 8%
5%


uAuAcuGuGuAAcAAucuATsT
718
uAGAUUGUuAcAcAGuAuATsT
719
AD-14153
17%
11% 


cuuAGuAGuGuccAGGAAATsT
720
UUUCCUGGAcACuACuAAGTsT
721
AD-14154
16%
4%


ucAGAuGGAcGuAAGGcAGTsT
722
CUGCCUuACGUCcAUCUGATsT
723
AD-14155
11%
1%


AGAuAAAuuGAuAGcAcAATsT
724
UUGUGCuAUcAAUUuAUCUTsT
725
AD-14156
10%
1%


cAAcAGGuAcGAcAccAcATsT
726
UGUGGUGUCGuACCUGUUGTsT
727
AD-14157
29%
3%


uGcAAuGuAAAuAcGuAuuTsT
728
AAuACGuAUUuAcAUUGcATsT
729
AD-14158
51%
3%


AGucAGAAuuuuAucuAGATsT
730
UCuAGAuAAAAUUCUGACUTsT
731
AD-14159
53%
5%


cuAGAAAucuuuuAAcAccTsT
732
GGUGUuAAAAGAUUUCuAGTsT
733
AD-14160
40%
3%


AAuAAAucuAAcccuAGuuTsT
734
AACuAGGGUuAGAUUuAUUTsT
735
AD-14161
83%
7%


AAuuuucuGcucAcGAuGATsT
736
UcAUCGUGAGcAGAAAAUUTsT
737
AD-14162
44%
6%


GcccucAGuAAAuccAuGGTsT
738
CcAUGGAUUuACUGAGGGCTsT
739
AD-14163
57%
3%


AcGuuuAAAAcGAGAucuuTsT
740
AAGAUCUCGUUUuAAACGUTsT
741
AD-14164
 4%
1%


AGGAGAuAGAAcGuuuAAATsT
742
UUuAAACGUUCuAUCUCCUTsT
743
AD-14165
11%
1%


GAccGucAuGGcGucGcAGTsT
744
CUGCGACGCcAUGACGGUCTsT
745
AD-14166
90%
5%


AccGucAuGGcGucGcAGcTsT
746
GCUGCGACGCcAUGACGGUTsT
747
AD-14167
49%
1%


GAAcGuuuAAAAcGAGAucTsT
748
GAUCUCGUUUuAAACGUUCTsT
749
AD-14168
12%
2%


uuGAGcuuAAcAuAGGuAATsT
750
UuACCuAUGUuAAGCUcAATsT
751
AD-14169
66%
4%


AcuAAAuuGAucucGuAGATsT
752
UCuACGAGAUcAAUUuAGUTsT
753
AD-14170
52%
6%


ucGuAGAAuuAucuuAAuATst
754
uAUuAAGAuAAUUCuACGATsT
755
AD-14171
42%
4%


GGAGAuAGAAcGuuuAAAATsT
756
UUUuAAACGUUCuAUCUCCTsT
757
AD-14172
 3%
1%


AcAAcuuAuuGGAGGuuGuTsT
758
AcAACCUCcAAuAAGUUGUTsT
759
AD-14173
29%
2%


uGAGcuuAAcAuAGGuAAATsT
760
UUuACCuAUGUuAAGCUcATsT
761
AD-14174
69%
2%


AucucGuAGAAuuAucuuATsT
762
uAAGAuAAUUCuACGAGAUTsT
763
AD-14175
53%
3%


cuGcGuGcAGucGGuccucTsT
764
GAGGACCGACUGcACGcAGTsT
765
AD-14176
111% 
4%


cAcGcAGcGcccGAGAGuATsT
766
uACUCUCGGGCGCUGCGUGTsT
767
AD-14177
87%
6%


AGuAccAGGGAGAcuccGGTsT
768
CCGGAGUCUCCCUGGuACUTsT
769
AD-14178
59%
2%


AcGGAGGAGAuAGAAcGuuTsT
770
AACGUUCuAUCUCCUCCGUTsT
771
AD-14179
 9%
2%


AGAAcGuuuAAAAcGAGAuTsT
772
AUCUCGUUUuAAACGUUCUTsT
773
AD-14180
43%
2%


AAcGuuuAAAAcGAGAucuTsT
774
AGAUCUCGUUUuAAACGUUTsT
775
AD-14181
70%
10% 


AGcuuGAGcuuAAcAuAGGTsT
776
CCuAUGUuAAGCUcAAGCUTsT
777
AD-14182
100% 
7%


AGcuuAAcAuAGGuAAAuATsT
778
uAUUuACCuAUGUuAAGCUTsT
779
AD-14183
60%
5%


uGAGAcuAcAAAAccuAucTsT
780
GAuAGGUUUUGuAGCUCuATsT
781
AD-14184
129% 
6%


uAGuuGuAuccuccuuuuATsT
782
uAAAGGAGGGAuAcAACuATsT
783
AD-14185
62%
4%


AccAcccAGAcAucuGAcuTsT
784
AGUcAGAUGUCUGGGUGGUTsT
785
AD-14186
42%
3%


AGAAAcuAAAuuGAucucGTsT
786
CGAGAUcAAUUuAGUUUCUTsT
787
AD-14187
123% 
12% 


ucucGuAGAAuuAucuuAATsT
788
UuAAGAuAAUUCuACGAGATsT
789
AD-14188
38%
2%


cAAcuuAuuGGAGGuuGuATsT
790
uAcAACCUCcAAuAAGUUGTsT
791
AD-14189
13%
1%


uuGuAucccuccuuuAAGuTsT
792
ACUuAAAGGAGGGAuAcAATsT
793
AD-14190
59%
3%


ucAcAAcuuAuuGGAGGuuTsT
794
AACCUCcAAuAAGUUGUGATsT
795
AD-14191
93%
3%


AGAAcuGuAcucuucucAGTsT
796
CUGAGAAGAGuAcAGUUCUTsT
797
AD-14192
45%
5%


GAGcuuAAcAuAGGuAAAuTsT
798
AUUuACCuAUGUuAAGCUCTsT
799
AD-14193
57%
3%


cAccAAcAucuGuccuuAGTsT
800
CuAAGGAcAGAUGUUGGUGTsT
801
AD-14194
51%
4%


AAAGcccAcuuuAGAGuAuTsT
802
AuACUCuAAAGUGGGCUUUTsT
803
AD-14195
77%
5%


AAGcccAcuuuAGAGuAuATsT
804
uAuACUCuAAAGUGGGCUUTsT
805
AD-14196
42%
6%


GAccuuAuuuGGuAAucuGTsT
806
cAGAUuACcAAAuAAGGUCTsT
807
AD-14197
15%
2%


GAuuAAcGuAcucAAGAcuTsT
808
AGUCUUGAGuAcAUuAAUCTsT
809
AD-14198
12%
2%


cuuuAAGAGGccuAAcucATsT
810
UGAGUuAGGCCUCUuAAAGTsT
811
AD-14199
18%
2%


uuAAAccAAAcccuAuuGATsT
812
UcAAuAGGGUUUGGUUuAATsT
813
AD-14200
72%
9%


ucuGuuGGAGAucuAuAAuTsT
814
AUuAuAGAUCUCcAAcAGATsT
815
AD-14201
 9%
3%


cuGAuGuuucuGAGAGAcuTsT
816
AGUCUCUcAGAAAcAUcAGTsT
817
AD-14202
25%
3%


GcAuAcucuAGucGuucccTsT
818
GGGAACGACuAGAGuAUGCTsT
819
AD-14203
21%
1%


GuuccuuAucGAGAAucuATsT
820
uAGAUUCUCGAuAAGGAACTsT
821
AD-14204
 4%
2%


GcAcuuGGAucucucAcAuTsT
822
AUGUGAGAGAUCcAAGUGCTsT
823
AD-14205
 5%
1%


AAAAAAGGAAcuAGAuGGcTsT
824
GCcAUCuAGUUCCUUUUUUTsT
825
AD-14206
79%
6%


AGAGcAGAuuAccucuGcGTsT
826
CGcAGAGGuAAUCUGCUCUTsT
827
AD-14207
55%
2%


AGcAGAuuAccucuGcGAGTsT
828
CUCGcAGAGGuAAUCUGCUTsT
829
AD-14208
100% 
4%


cccuGAcAGAGuucAcAAATsT
830
UUUGUGAACUCUGUcAGGGTsT
831
AD-14209
34%
3%


GuuuAccGAAGuGuuGuuuTsT
832
AAAcAAcACUUCGGuAAACTsT
833
AD-14210
13%
2%


uuAcAGuAcAcAAcAAGGATsT
834
UCCUUGUUGUGuACUGuAATsT
835
AD-14211
 9%
1%


AcuGGAucGuAAGAAGGcATsT
836
UGCCUUCUuACGAUCcAGUTsT
837
AD-14212
20%
3%


GAGcAGAuuAccucuGcGATsT
838
UCGcAGAGGuAAUCUGCUCTsT
839
AD-14213
48%
5%


AAAAGAAGuuAGuGuAcGATsT
840
UCGuAcACuAACUUCUUUUTsT
841
AD-14214
28%
18% 


GAccAuuuAAuuuGGcAGATsT
842
UCUGCcAAAUuAAAUGGUCTsT
843
AD-14215
132% 
0%


GAGAGGAGuGAuAAuuAAATsT
844
UUuAAUuAUcACUCCUCUCTsT
845
AD-14216
 3%
0%


cuGGAGGAuuGGcuGAcAATsT
846
UUGUcAGCcAAUCCUCcAGTsT
847
AD-14217
19%
1%


cucuAGucGuucccAcucATsT
848
UGAGUGGGAACGACuAGAGTsT
849
AD-14218
67%
8%


GAuAccAuuAcuAcAGuAGTsT
850
CuACUGuAGuAAUGGuAUCTsT
851
AD-14219
76%
4%


uucGucuGcGAAGAAGAAATsT
852
UUUCUUCUUCGcAGACGAATsT
853
AD-14220
33%
8%


GAAAAGAAGuuAGuGuAcGTsT
854
CGuAcACuAACUUCUUUUCTsT
855
AD-14221
25%
2%


uGAuGuuuAccGAAGuGuuTsT
856
AAcACUUCGGuAAAcAUcATsT
857
AD-14222
 7%
2%


uGuuuGuccAAuucuGGAuTsT
858
AUCcAGAAUUGGAcAAAcATsT
859
AD-14223
19%
2%


AuGAAGAGuAuAccuGGGATsT
860
UCCcAGGuAuACUCUUcAUTsT
861
AD-14224
13%
1%


GcuAcucuGAuGAAuGcAuTsT
862
AUGcAUUcAUcAGAGuAGCTsT
863
AD-14225
15%
2%


GcccuuGuAGAAAGAAcAcTsT
864
GUGUUCUUUCuAcAAGGGCTsT
865
AD-14226
11%
0%


ucAuGuuccuuAucGAGAATsT
866
UUCUCGAuAAGGAAcAUGATsT
867
AD-14227
5%
1%


GAAuAGGGuuAcAGAGuuGTsT
868
cAACUCUGuAACCCuAUUCTsT
869
AD-14228
34%
3%


cAAAcuGGAucGuAAGAAGTsT
870
CUUCUuACGAUCcAGUUUGTsT
871
AD-14229
15%
2%


cuuAuuuGGuAAucuGcuGTsT
872
cAGcAGAUuACcAAAuAAGTsT
873
AD-14230
20%
1%


AGcAAuGuGGAAAccuAAcTsT
874
GUuAGGUUUCcAcAUUGCUTsT
875
AD-14231
18%
1%


AcAAuAAAGcAGAcccAuuTsT
876
AAUGGGUCUGCUUuAUUGUTsT
877
AD-14232
21%
1%


AAccAcuuAGuAGuGuccATsT
878
UGGAcACuACuAAGUGGUUTsT
879
AD-14233
106% 
12% 


AGucAAGAGccAucuGuAGTsT
880
CuAcAGAUGGCUCUUGACUTsT
881
AD-14234
35%
3%


cucccuAGAcuucccuAuuTsT
882
AAuAGGGAAGUCuAGGGAGTsT
883
AD-14235
45%
4%


AuAGcuAAAuuAAAccAAATsT
884
UUUGGUUuAAUUuAGCuAUTsT
885
AD-14236
23%
3%


uGGcuGGuAuAAuuccAcGTsT
886
CGUGGAAUuAuACcAGCcATsT
887
AD-14237
79%
9%


uuAuuuGGuAAucuGcuGuTsT
888
AcAGcAGAUuACcAAAuAATsT
889
AD-14238
92%
7%


AAcuAGAuGGcuuucucAGTsT
890
CUGAGAAAGCcAUCuAGUUTsT
891
AD-14239
20%
2%


ucAuGGcGucGcAGccAAATsT
892
UUUGGCUGCGACGCcAUGATsT
893
AD-14240
71%
6%


AcuGGAGGAuuGGcuGAcATsT
894
UGUcAGCcAAUCCUCcAGUTsT
895
AD-14241
14%
1%


cuAuAAuuGcAcuAucuuuTsT
896
AAAGAuAGUGcAAUuAuAGTsT
897
AD-14242
11%
2%


AAAGGucAccuAAuGAAGATsT
898
UCUUcAUuAGGUGACCUUUTsT
899
AD-14243
11%
1%


AuGAAuGcAuAcucuAGucTsT
900
GACuAGAGuAUGcAUUcAUTsT
901
AD-14244
15%
2%


AAcAuAuuGAAuAAGccuGTsT
902
cAGGCUuAUUcAAuAUGUUTsT
903
AD-14245
80%
7%


AAGAAGGcAGuuGAccAAcTsT
904
GUUGGUcAACAGCCUUCUUTsT
905
AD-14246
57%
5%


GAuAcuAAAAGAAcAAucATsT
906
UGAUUGUUCUUUuAGuAUCTsT
907
AD-14247
 9%
3%


AuAcuGAAAAucAAuAGucTsT
908
GACuAUUGAUUUUcAGuAUTsT
909
AD-14248
39%
4%


AAAAAGGAAcuAGAuGGcuTsT
910
AGCcAUCuAGUUCCUUUUUTsT
911
AD-14249
64%
2%


GAAcuAGAuGGcuuucucATsT
912
UGAGAAAGCcAUCuAGUUCTsT
913
AD-14250
18%
2%


GAAAccuAAcuGAAGAccuTsT
914
AGGUCUUcAGUuAGGUUUCTsT
915
AD-14251
56%
6%


uAcccAucAAcAcuGGuAATsT
916
UuACcAGUGUUGAUGGGuATsT
917
AD-14252
48%
6%


AuuuuGAuAucuAcccAuuTsT
918
AAUGGGuAGAuAUcAAAAUTsT
919
AD-14253
39%
5%


AucccuAuAGuucAcuuuGTsT
920
cAAAGUGAACuAuAGGGAUTsT
921
AD-14254
44%
8%


AuGGGcuAuAAuuGcAcuATsT
922
uAGUGcAAUuAuAGCCcAUTsT
923
AD-14255
108% 
8%


AGAuuAccucuGcGAGcccTsT
924
GGGCUCGcAGAGGuAAUCUTsT
925
AD-14256
108% 
6%


uAAuuccAcGuAcccuucATsT
926
UGAAGGGuACGUGGAAUuATsT
927
AD-14257
23%
2%


GucGuucccAcucAGuuuuTsT
928
AAAACuGAGuGGGAACGACTsT
929
AD-14258
21%
3%


AAAucAAucccuGuuGAcuTsT
930
AGUcAAcAGGGAUUGAUUUTsT
931
AD-14259
19%
2%


ucAuAGAGcAAAGAAcAuATsT
932
uAUGUUCUUUGCUCuAUGATsT
933
AD-14260
10%
1%


uuAcuAcAGuAGcAcuuGGTsT
934
CcAAGUGCuACUGuAGuAATsT
935
AD-14261
76%
3%


AuGuGGAAAccuAAcuGAATsT
936
UUcAGUuAGGUUUCcAcAUTsT
937
AD-14262
13%
2%


uGuGGAAAccuAAcuGAAGTsT
938
CUUcAGUuAGGUUUCcAcATsT
939
AD-14263
14%
2%


uGuuccuuAAAuGAAAGGGTsT
940
CCCUUUcAUUuAAGGAAGATsT
941
AD-14264
65%
3%


uGAAGAAccucuAAGucAATsT
942
UUGACUuAGAGGUUCUUcATsT
943
AD-14265
13%
1%


AGAGGucuAAAGuGGAAGATsT
944
UCUUCcACUUuAGACCUCUTsT
945
AD-14266
18%
3%


AuAucuAcccAuuuuucuGTsT
946
cAGAAAAAUGGGuAGAuAUTsT
947
AD-14267
50%
9%


uAAGccuGAAGuGAAucAGTsT
948
CUGAUUcACUUcAGGCUuATsT
949
AD-14268
13%
3%


AGAuGcAGAccAuuuAAuuTsT
950
AAUuAAAUGGUCUGcAUCUTsT
951
AD-14269
19%
4%


AGuGuuGuuuGuccAAuucTsT
952
GAAUUGGAcAAAcAAcACUTsT
953
AD-14270
11%
2%


cuAuAAuGAAGAGcuuuuuTsT
954
AAAAAGCUCUUcAUuAuAGTsT
955
AD-14271
11%
1%


AGAGGAGuGAuAAuuAAAGTsT
956
CUUuAAUuAUcACUCCUCUTsT
957
AD-14272
 7%
1%


uuucucuGuuAcAAuAcAuTsT
958
AUGuAUUGuAAcAGAGAAATsT
959
AD-14273
14%
2%


AAcAucuAuAAuuGcAAcATsT
960
UGUUGcAAUuAuAGAUGUUTsT
961
AD-14274
73%
4%


uGcuAGAAGuAcAuAAGAcTsT
962
GUCUuAUGuACUUCuAGcATsT
963
AD-14275
10%
1%


AAuGuAcucAAGAcuGAucTsT
964
GAUcAGUCUUGACuAcAUUTsT
965
AD-14276
89%
2%


GuAcucAAGAcuGAucuucTsT
966
GAAGAUcAGUCUUGAGuACTsT
967
AD-14277
 7%
1%


cAcucuGAuAAAcucAAuGTsT
968
cAUUGAGUUuAUcAGAGUGTsT
969
AD-14278
12%
1%


AAGAGcAGAuuAccucuGcTsT
970
GcAGAGGuAAUCUGCUCUUTsT
971
AD-14279
104% 
3%


ucuGcGAGcccAGAucAAcTsT
972
GUUGAUCUGGGCUCGcAGATsT
973
AD-14280
21%
2%


AAcuuGAGccuuGuGuAuATsT
974
uAuAcAcAAGGCUcAAGUUTsT
975
AD-14281
43%
3%


GAAuAuAuAuAucAGccGGTsT
976
CCGGCUGAuAuAuAuAUUCTsT
977
AD-14282
45%
6%


uGucAucccuAuAGuucAcTsT
978
GUGAACuAuAGGGAUGAcATsT
979
AD-14283
35%
5%


GAucuGGcAAccAuAuuucTsT
980
GAAAuAUGGUUGCcAGAUCTsT
981
AD-14284
58%
3%


uGGcAAccAuAuuucuGGATsT
982
UCcAGAAAuAUGGUUGCcATsT
983
AD-14285
48%
3%


GAuGuuuAccGAAGuGuuGTsT
984
cAAcACUUCGGuAAAcAUCTsT
985
AD-14286
49%
3%


uuccuuAucGAGAAucuAATsT
986
UuAGAUUCUCGAuAAGGAATsT
987
AD-14287
6%
1%


AGcuuAAuuGcuuucuGGATsT
988
UCcAGAAAGcAAUuAAGCUTsT
989
AD-14288
50%
2%


uuGcuAuuAuGGGAGAccATsT
990
UGGUCUCCcAuAAuAGcAATsT
991
AD-14289
48%
1%


GucAuGGcGucGcAGccAATsT
992
UUGGCUGCGACGCcAUGACTsT
993
AD-14290
112% 
7%


uAAuuGcAcuAucuuuGcGTsT
994
CGcAAAGAuAGUGcAAUuATsT
995
AD-14291
77%
2%


cuAucuuuGcGuAuGGccATsT
996
UGGCcAuACGcAAAGAuAGTsT
997
AD-14292
80%
6%


ucccuAuAGuucAcuuuGuTsT
998
AcAAAGUGAACuAuAGGGATsT
999
AD-14293
58%
2%


ucAAccuuuAAuucAcuuGTsT
1000
cAAGUGAAUuAAAGGUUGATsT
1001
AD-14294
77%
2%


GGcAAccAuAuuucuGGAATsT
1002
UUCcAGAAAuAUGGUUGCCTsT
1003
AD-14295
62%
2%


AuGuAcucAAGAcuGAucuTsT
1004
AGAUcAGUCUUGAGuAcAUTsT
1005
AD-14296
59%
4%


GcAGAccAuuuAAuuuGGcTsT
1006
GCcAAAUuAAAUGGUCUGCTsT
1007
AD-14297
37%
1%


ucuGAGAGAcuAcAGAuGuTsT
1008
AcAUCUGuAGUCUCUcAGATsT
1009
AD-14298
21%
1%


uGcucAuAGAGcAAAGAAcTsT
1010
GUUCUUUGCUCuAUGAGcATsT
1011
AD-14299
 6%
1%


AcAuAAGAccuuAuuuGGuTsT
1012
ACcAAAuAAGGUCUuAUGUTsT
1013
AD-14300
17%
2%


uuuGuGcuGAuucuGAuGGTsT
1014
CcAUcAGAAUcAGcAcAAATsT
1015
AD-14301
97%
6%


ccAucAAcAcuGGuAAGAATsT
1016
UUCUuACcAGUGUUGAUGGTsT
1017
AD-14302
13%
1%


AGAcAAuuccGGAuGuGGATsT
1018
UCcAcAUCCGGAAUUGUCUTsT
1019
AD-14303
13%
3%


GAAcuuGAGccuuGuGuAuTsT
1020
AuAcAcAAGGCUcAAGUUCTsT
1021
AD-14304
38%
2%


uAAuuuGGcAGAGcGGAAATsT
1022
UUUCCGCUCUGCcAAAUuATsT
1023
AD-14305
14%
2%


uGGAuGAAGuuAuuAuGGGTsT
1024
CCcAuAAuAACUUcAUCcATsT
1025
AD-14306
22%
4%


AucuAcAuGAAcuAcAAGATsT
1026
UCUUGuAGUUcAUGuAGAUTsT
1027
AD-14307
26%
6%


GGuAuuuuuGAucuGGcAATsT
1028
UUGCcAGAUcAAAAAuACCTsT
1029
AD-14308
62%
8%


cuAAuGAAGAGuAuAccuGTsT
1030
cAGGuAuACUCUUcAUuAGTsT
1031
AD-14309
52%
5%


uuuGAGAAAcuuAcuGAuATsT
1032
uAUcAGuAAGUUUCUcAAATsT
1033
AD-14310
32%
3%


cGAuAAGAuAGAAGAucAATsT
1034
UUCAUCUUCuAUCUuAUCGTsT
1035
AD-14311
23%
2%


cuGGcAAccAuAuuucuGGTsT
1036
CcAGAAAuAUGGUUGCcAGTsT
1037
AD-14312
49%
6%


uAGAuAccAuuAcuAcAGuTsT
1038
ACUGUAGuAAUGGuAUCuATsT
1039
AD-14313
69%
4%


GuAuuAAAuuGGGuuucAuTsT
1040
AUGAAACCcAAUUuAAuACTsT
1041
AD-14314
52%
3%


AAGAccuuAuuuGGuAAucTsT
1042
GAUuACcAAAuAAGGUCUUTsT
1043
AD-14315
66%
4%


GcuGuuGAuAAGAGAGcucTsT
1044
GAGCUCUCUuAUcAAcAGCTsT
1045
AD-14316
19%
4%


uAcucAuGuuucucAGAuuTsT
1046
AAUCUGAGAAAcAUGAGuATsT
1047
AD-14317
16%
5%


cAGAuGGAcGuAAGGcAGcTsT
1048
GCUGCCUuACGUCcAUCUGTsT
1049
AD-14318
52%
11% 


uAucccAAcAGGuAcGAcATsT
1050
UGUCGuACCUGUUGGGAuATsT
1051
AD-14319
28%
11% 


cAuuGcuAuuAuGGGAGAcTsT
1052
GUCUCCcAuAAuAGcAAUGTsT
1053
AD-14320
52%
10% 


cccucAGuAAAuccAuGGuTsT
1054
ACcAUGGAUUuACUGAGGGTsT
1055
AD-14321
53%
6%


GGucAuuAcuGcccuuGuATsT
1056
uAcAAGGGcAGuAAUGACCTsT
1057
AD-14322
20%
2%


AAccAcucAAAAAcAuuuGTsT
1058
cAAAUGUUUUUGAGUGGUUTsT
1059
AD-14323
116% 
6%


uuuGcAAGuuAAuGAAucuTsT
1060
AGAUUcAUuAACUUGcAAATsT
1061
AD-14324
14%
2%


uuAuuuucAGuAGucAGAATsT
1062
UUCUGACuACUGAAAAuAATsT
1063
AD-14325
50%
2%


uuuucucGAuucAAAucuuTsT
1064
AAGAUUuGAAUCGAGAAAATsT
1065
AD-14326
47%
3%


GuAcGAAAAGAAGuuAGuGTsT
1066
cACuAACUUCUUUUCGuACTsT
1067
AD-14327
18%
2%


uuuAAAAcGAGAucuuGcuTsT
1068
AGcAAGAUCUCGUUUuAAATsT
1069
AD-14328
19%
1%


GAAuuGAuuAAuGuAcucATsT
1070
UGAGuAcAUuAAUcAAUUCTsT
1071
AD-14329
94%
10% 


GAuGGAcGuAAGGcAGcucTsT
1072
GAGCUGCCUuACGUCcAUCTsT
1073
AD-14330
60%
4%


cAucuGAcuAAuGGcucuGTsT
1074
cAGAGCcAUuAGUcAGAUGTsT
1075
AD-14331
54%
7%


GuGAuccuGuAcGAAAAGATsT
1076
UCUUUUCGuAcAGGAUcACTsT
1077
AD-14332
22%
4%


AGcucuuAuuAAGGAGuAuTsT
1078
AuACUCCUuAAuAAGAGCUTsT
1079
AD-14333
70%
10% 


GcucuuAuuAAGGAGuAuATsT
1080
uAuACUCCUuAAuAAGAGCTsT
1081
AD-14334
18%
3%


ucuuAuuAAGGAGuAuAcGTsT
1082
CGuAuACUCCUuAAuAAGATsT
1083
AD-14335
38%
6%


uAuuAAGGAGuAuAcGGAGTsT
1084
CUCCGuAuACUCCUuAAuATsT
1085
AD-14336
16%
3%


cuGcAGcccGuGAGAAAAATsT
1086
UUUUUCUcACGGGCUGcAGTsT
1087
AD-14337
65%
4%


ucAAGAcuGAucuucuAAGTsT
1088
CUuAGAAGAUcAGUCUUGATsT
1089
AD-14338
18%
0%


cuucuAAGuucAcuGGAAATsT
1090
UUUCcAGUGAACUuAGAAGTsT
1091
AD-14339
20%
4%


uGcAAGuuAAuGAAucuuuTsT
1092
AAAGAUUcAUuAACUUGcATsT
1093
AD-14340
24%
1%


AAucuAAGGAuAuAGucAATsT
1094
UUGACuAuAUCCUuAGAUUTsT
1095
AD-14341
27%
3%


AucucuGAAcAcAAGAAcATsT
1096
UGUUCUUGUGUUcAGAGAUTsT
1097
AD-14342
13%
1%


uucuGAAcAGuGGGuAucuTsT
1098
AGAuACCcACUGUUcAGAATsT
1098
AD-14343
19%
1%


AGuuAuuuAuAcccAucAATsT
1100
UUGAUGGGuAuAAAuAACUTsT
1101
AD-14344
23%
2%


AuGcuAAAcuGuucAGAAATsT
1102
UUUCUGAAcAGUUuAGcAUTsT
1103
AD-14345
21%
4%


cuAcAGAGcAcuuGGuuAcTsT
1104
GuAACcAAGUGCUCUGuAGTsT
1105
AD-14346
18%
2%


uAuAuAucAGccGGGcGcGTsT
1106
CGCGCCCGGCUGAuAuAuATsT
1107
AD-14347
67%
2%


AuGuAAAuAcGuAuuucuATsT
1108
uAGAAAuACGuAUUuAcAUTsT
1109
AD-14348
39%
3%


uuuuucucGAuucAAAucuTsT
1110
AGAUUuGAAUCGAGAAAAATsT
1111
AD-14349
83%
6%


AAucuuAAcccuuAGGAcuTsT
1112
AGUCCuAAGGGUuAAGAUUTsT
1113
AD-14350
54%
2%


ccuuAGGAcucuGGuAuuuTsT
1114
AAAuACcAGAGUCCuAAGGTsT
1115
AD-14351
57%
8%


AAuAAAcuGcccucAGuAATsT
1116
UuACUGAGGGcAGUUuAUUTsT
1117
AD-14352
82%
3%


GAuccuGuAcGAAAAGAAGTsT
1118
CUUCUUUUCGuAcAGGAUCTsT
1119
AD-14353
 2%
1%


AAuGuGAuccuGuAcGAAATsT
1120
UUUCGuAcAGGAUcAcAUUTsT
1121
AD-14354
18%
11% 


GuGAAAAcAUUGGccGuucTsT
1122
GAACGGCcAAUGUUUUcACTsT
1123
AD-14355
 2%
1%


cuuGAGGAAAcucuGAGuATsT
1124
uACUcAGAGUUUCCUcAAGTsT
1125
AD-14356
 8%
2%


cGuuuAAAAcGAGAucuuGTsT
1126
cAAGAUCUCGUUUuAAACGTsT
1127
AD-14357
 6%
3%


uuAAAAcGAGAucuuGcuGTsT
1128
cAGcAAGAUCUCGUUUuAATsT
1129
AD-14358
98%
17% 


AAAGAuGuAucuGGucuccTsT
1130
GGAGACcAGAuAcAUCUUUTsT
1131
AD-14359
10%
1%


cAGAAAAuGuGucuAcucATsT
1132
UGAGuAGAcAcAUUUUCUGTsT
1133
AD-14360
 6%
4%


cAGGAAuuGAuuAAuGuAcTsT
1134
GuAcAUuAAUcAAUUCCUGTsT
1135
AD-14361
30%
5%


AGucAAcuAAAGcAuAuuuTsT
1136
AAAuAUGCUUuAGUUGACUTsT
1137
AD-14362
20%
2%


uGuGuAAcAAucuAcAuGATsT
1138
UcAUGuAGAUUGUuAcAcATsT
1139
AD-14363
60%
6%


AuAccAuuuGuuccuuGGuTsT
1140
ACcAAGGAAcAAAUGGuAUTsT
1141
AD-14364
12%
9%


GcAGAAAucuAAGGAuAuATsT
1142
uAuAUCCUuAGAUUUCUGCTsT
1143
AD-14365
 5%
2%


uGGcuucucAcAGGAAcucTsT
1144
GAGUUCCUGUGAGAAGCcATsT
1145
AD-14366
28%
5%


GAGAuGuGAAucucuGAAcTsT
1146
GUUcAGAGAUUcAcAUCUCTsT
1147
AD-14367
42%
4%


uGuAAGccAAuGuuGuGAGTsT
1148
CUcAcAAcAUUGGCUuAcATsT
1149
AD-14368
93%
12% 


AGccAAuGuuGuGAGGcuuTsT
1150
AAGCCUcAcAAcAUUGGCUTsT
1151
AD-14369
65%
4%


uuGuGAGGcuucAAGuucATsT
1152
UGAACUUGAAGCCUcAcAATsT
1153
AD-14370
 5%
2%


AGGcAGcucAuGAGAAAcATsT
1154
UGUUUCUcAUGAGCUGCCUTsT
1155
AD-14371
54%
5%


AuAAAuuGAuAGcAcAAAATsT
1156
UUUUGUGCuAUcAAUUuAUTsT
1157
AD-14372
 4%
1%


AcAAAAucuAGAAcuuAAuTsT
1158
AUuAAGUUCuAGAUUUUGUTsT
1159
AD-14373
 5%
1%


GAuAucccAAcAGGuAcGATsT
1160
UCGuACCUGUUGGGAuAUCTsT
1161
AD-14374
92%
6%


AAGuuAuuuAuAcccAucATsT
1162
UGAUGGGuAuAAAuAACUUTsT
1163
AD-14375
76%
4%


uGuAAAuAcGuAuuucuAGTsT
1164
CuAGAAAuACGuAUUuAcATsT
1165
AD-14376
70%
5%


ucuAGuuuucAuAuAAAGuTsT
1166
ACUUuAuAUGAAAACuAGATsT
1167
AD-14377
48%
4%


AuAAAGuAGuucuuuuAuATsT
1168
uAuAAAAGAACuACUUuAUTsT
1169
AD-14378
48%
3%


ccAuuuGuAGAGcuAcAAATsT
1170
UUUGuAGCUCuAcAAAUGGTsT
1171
AD-14379
44%
5%


uAuuuucAGuAGucAGAAuTsT
1172
AUUCUGACuACUGAAAAuATsT
1173
AD-14380
35%
16% 


AAAucuAAcccuAGuuGuATsT
1174
uAcAACuAGGGUuAGAUUUTsT
1175
AD-14381
44%
5%


cuuuAGAGuAuAcAuuGcuTsT
1176
AGcAAUGuAuACUCuAAAGTsT
1177
AD-14382
28%
1%


AucuGAcuAAuGGcucuGuTsT
1178
AcAGAGCcAUuAGUcAGAUTsT
1179
AD-14383
55%
11% 


cAcAAuGAuuuAAGGAcuGTsT
1180
cAGUCCUuAAAUcAUUGUGTsT
1181
AD-14384
48%
9%


ucuuuuucucGAuucAAAuTsT
1182
AUUuGAAUCGAGAAAAAGATsT
1183
AD-14385
36%
2%


cuuuuucucGAuucAAAucTsT
1184
GAUUuGAAUCGAGAAAAAGTsT
1185
AD-14386
41%
7%


AuuuucuGcucAcGAuGAGTsT
1186
CUcAUCGUGAGcAGAAAAUTsT
1187
AD-14387
38%
3%


uuucuGcucAcGAuGAGuuTsT
1188
AACUcAUCGUGAGcAGAAATsT
1189
AD-14388
50%
4%


AGAGcuAcAAAAccuAuccTsT
1190
GGAuAGGUUUUGuAGCUCUTsT
1191
AD-14389
98%
6%


GAGccAAAGGuAcAccAcuTsT
1192
AGUGGUGuACCUUUGGCUCTsT
1193
AD-14390
43%
0%


GccAAAGGuAcAccAcuAcTsT
1194
GuAGUGGUGuACCUUUGGCTsT
1195
AD-14391
48%
4%


GAAcuGuAcucuucucAGcTsT
1196
GCUGAGAAGAGuAcAGUUCTsT
1197
AD-14392
44%
3%


AGGuAAAuAucAccAAcAuTsT
1198
AUGUUGGUGAuAUUuACCUTsT
1199
AD-14393
37%
2%


AGcuAcAAAAccuAuccuuTsT
1200
AAGGAuAGGUUUUGuAGCUTsT
1201
AD-14394
114% 
7%


uGuGAAAGcAuuuAAuuccTsT
1202
GGAAUuAAAUGCUUUcAcATsT
1203
AD-14395
55%
4%


GcccAcuuuAGAGuAuAcATsT
1204
UGuAuACUCuAAAGUGGGCTsT
1205
AD-14396
49%
5%


uGuGccAcAcuccAAGAccTsT
1206
GGUCUUGGAGUGUGGcAcATsT
1207
AD-14397
71%
6%


AAAcuAAAuuGAucucGuATsT
1208
uACGAGAUcAAUUuAGUUUTsT
1209
AD-14398
81%
7%


uGAucucGuAGAAuuAucuTsT
1210
AGAuAAUUCuACGAGAUcATsT
1211
AD-14399
38%
4%


GcGuGcAGucGGuccuccATsT
1212
UGGAGGACCGACUGcACGCTsT
1213
AD-14400
106% 
8%


AAAGuuuAGAGAcAucuGATsT
1214
UcAGAUGUCUCuAAACUUUTsT
1215
AD-14401
47%
3%


cAGAAGGAAuAuGuAcAAATsT
1216
UUUGuAcAuAUUCCUUCUGTsT
1217
AD-14402
31%
1%


cGcccGAGAGuAccAGGGATsT
1218
UCCCUGGuACUCUCGGGCGTsT
1219
AD-14403
105% 
4%


cGGAGGAGAuAGAAcGuuuTsT
1220
AAACGUUCuAUCUCCUCCGTsT
1221
AD-14404
 3%
1%


AGAuAGAAcGuuuAAAAcGTsT
1222
CGUUUuAAACGUUCuAUCUTsT
1223
AD-14405
15%
1%


GGAAcAGGAAcuucAcAAcTsT
1224
GUuGuGAAGUUCCuGUUCCTsT
1225
AD-14406
44%
5%


GuGAGccAAAGGuAcAccATsT
1226
UGGUGuACCUUUGGCUcACTsT
1227
AD-14407
41%
4%


AuccucccuAGAcuucccuTsT
1228
AGGGAAGUCuAGGGAGGAUTsT
1229
AD-14408
104% 
3%


cAcAcuccAAGAccuGuGcTsT
1230
GcAcAGGUCUUGGAGUGUGTsT
1231
AD-14409
67%
4%


AcAGAAGGAAuAuGuAcAATsT
1232
UUGuAcAuAUUCCUUCUGUTsT
1233
AD-14410
22%
1%


uuAGAGAcAUCUGAcuuuGTsT
1234
cAAAGUcAGAUGUCUCuAATsT
1235
AD-14411
29%
3%


AAuuGAucucGuAGAAuuATsT
1236
uAAUUCuACGAGAUcAAUUTsT
1237
AD-14412
31%
4%








Claims
  • 1. A double-stranded ribonucleic acid (dsRNA) for inhibiting the expression of a human kinesin family member 11 (Eg5) gene in a cell, wherein said dsRNA comprises a sense strand comprising a first sequence and an antisense strand comprising a second sequence complementary to SEQ ID NO:1311, wherein said first sequence is complementary to said second sequence and wherein said dsRNA is between 15 and 30 base pairs in length.
  • 2. The dsRNA of claim 1, wherein said dsRNA comprises at least one modified nucleotide.
  • 3. The dsRNA of claim 2, wherein said modified nucleotide is chosen from the group of: a 2′-O-methyl modified nucleotide, a nucleotide comprising a 5′-phosphorothioate group, and a terminal nucleotide linked to a cholesteryl derivative or dodecanoic acid bisdecylamide group.
  • 4. The dsRNA of claim 2, wherein said modified nucleotide is chosen from the group of: a 2′-deoxy-2′-fluoro modified nucleotide, a 2′-deoxy-modified nucleotide, a locked nucleotide, an abasic nucleotide, 2′-amino-modified nucleotide, 2′-alkyl-modified nucleotide, morpholino nucleotide, a phosphoramidate, and a non-natural base comprising nucleotide.
  • 5. An isolated cell comprising the dsRNA of claim 1.
  • 6. A pharmaceutical composition for inhibiting the expression of the Eg5 gene comprising the dsRNA of claim 1.
  • 7. The pharmaceutical composition of claim 6, further comprising a second dsRNA that inhibits the expression of the VEGF gene wherein the second dsRNA comprises a sense strand consisting of the sequence of SEQ ID NO:1242, and an antisense strand consisting of the sequence of SEQ ID NO:1243.
  • 8. A method for inhibiting expression of a human kinesin family member 11 (Eg5) gene in a cell, the method comprising: (a) introducing into the cell the dsRNA of claim 1; and(b) maintaining the cell produced in step (a) for a time sufficient to obtain degradation of the mRNA transcript of the Eg5 gene, thereby inhibiting expression of the Eg5 gene in the cell.
  • 9. The method of claim 8, wherein a second dsRNA that inhibits the expression of VEGF is introduced into said cell wherein the second dsRNA comprises a sense strand consisting of the sequence of SEQ ID NO:1242, and an antisense strand consisting of the sequence of SEQ ID NO:1243.
  • 10. A method of treating or managing pathological processes mediated by human kinesin family member 11 (Eg5) expression comprising administering to a patient in need of such treatment or management a therapeutically effective amount of the dsRNA of claim 1.
  • 11. The method of claim 10 further comprising administering a second dsRNA that inhibits the expression of VEGF wherein the second dsRNA comprises a sense strand consisting of the sequence of SEQ ID NO:1242, and an antisense strand consisting of the sequence of SEQ ID NO:1243.
  • 12. A vector for inhibiting the expression of a human kinesin family member 11 (Eg5) gene in a cell, said vector comprising a regulatory sequence operably linked to a nucleotide sequence that encodes at least one strand of a dsRNA, wherein one of the strands of said dsRNA is substantially complementary to SEQ ID NO:1311 and wherein said dsRNA is less than 30 base pairs in length.
  • 13. An isolated cell comprising the vector of claim 12.
  • 14. The dsRNA of claim 1, wherein said dsRNA, upon contact with a cell expressing said Eg5, inhibits expression of said Eg5 gene by at least 40%.
  • 15. The dsRNA of claim 1, wherein said dsRNA is 19-21 base pairs in length.
  • 16. The vector of claim 12, wherein said dsRNA, upon contact with a cell expressing said Eg5, inhibits expression of said Eg5 gene by at least 40%.
  • 17. The vector of claim 12, wherein said dsRNA is 19-21 base pairs in length.
  • 18. The pharmaceutical composition of claim 6, wherein the dsRNA is duplex AD12115 consisting of a sense strand consisting of SEQ ID NO:135 and an antisense strand consisting of SEQ ID NO:136.
  • 19. The pharmaceutical composition of claim 7, wherein the dsRNA of claim 1 is duplex AD12115 consisting of a sense strand consisting of SEQ ID NO:135 and an antisense strand consisting of SEQ ID NO:136.
  • 20. The method of claim 9, wherein said second dsRNA comprises a sense strand consisting of the sequence of SEQ ID NO:1242, and an antisense strand consisting of the sequence of SEQ ID NO:1243.
  • 21. The dsRNA of claim 1, wherein the dsRNA is duplex AD12115 consisting of a sense strand consisting of SEQ ID NO:135 and an antisense strand consisting of SEQ ID NO:136.
  • 22. The dsRNA of claim 21, wherein each strand is modified as follows to include a 2′-O-methyl ribonucleotide as indicated by a lower case letter “c” or “u” and a phosphorothioate as indicated by a lower case letter “s”: SEQ ID NO:135 is ucGAGAAucuAAAcuAAcuTsTSEQ ID NO:136 is AGUuAGUUuAGAUUCUCGATsT.
  • 23. A composition comprising the dsRNA of claim 1 and a second dsRNA that inhibits the expression of the VEGF gene, wherein the second dsRNA that inhibits the expression of VEGF comprises a sense strand consisting of the sequence of SEQ ID NO:1242, and an antisense strand consisting of the sequence of SEQ ID NO:1243.
  • 24. The composition of claim 22, wherein the dsRNA of claim 1 is duplex AD12115 consisting of a sense strand consisting of SEQ ID NO:135 and an antisense strand consisting of SEQ ID NO:136.
  • 25. The composition of claim 23, wherein the sense strand and antisense strand of the dsRNA of claim 1 are modified as follows to include a 2′-O-methyl ribonucleotide as indicated by a lower case letter “c” or “u” and a phosphorothioate as indicated by a lower case letter “s”: SEQ ID NO:135 is ucGAGAAucuAAAcuAAcuTsTSEQ ID NO:136 is AGUuAGUUuAGAUUCUCGATsT.
  • 26. The composition of claim 22 wherein the sense strand and antisense strand of the second dsRNA that inhibits the expression of VEGF are modified as follows to include a 2′-O-methyl ribonucleotide as indicated by a lower case letter “c” or “u” and a phosphorothioate as indicated by a lower case letter “s”: GcAcAuAGGAGAGAuGAGCUsU (SEQ ID NO:1242)AAGCUcAUCUCUCCuAuGuGCusG (SEQ ID NO:1243).
  • 27. The composition of claim 22, wherein the dsRNA of claim 1 is duplex AD12115 consisting of a sense strand consisting of SEQ ID NO:135 and an antisense strand consisting of SEQ ID NO:136 and the second dsRNA that inhibits the expression of VEGF comprises a sense strand consisting of the sequence of SEQ ID NO:1242 and an antisense strand consisting of the sequence of SEQ ID NO:1243.
  • 28. The composition of claim 26, wherein the sense and antisense strands of the dsRNA of claim 1 and the sense strand and antisense strand of the second dsRNA that inhibits the expression of VEGF are modified as follows to include a 2′-O-methyl ribonucleotide as indicated by a lower case letter “c” or “u” and a phosphorothioate as indicated by a lower case letter “s”: SEQ ID NO:135 is ucGAGAAucuAAAcuAAcuTsTSEQ ID NO:136 is AGUuAGUUuAGAUUCUCGATsTGcAcAuAGGAGAGAuGAGCUsU (SEQ ID NO:1242)AAGCUcAUCUCUCCuAuGuGCusG (SEQ ID NO:1243).
  • 29. A pharmaceutical composition comprising the composition of claim 22.
  • 30. At least one vector for inhibiting the expression of a human kinesin family member 11 (Eg5) gene and a VEGF gene in a cell, said vector comprising a regulatory sequence operably linked to a nucleotide sequence that encodes at least one strand of a first dsRNA and at least one strand of a second dsRNA, wherein one strand of said first dsRNA is substantially complementary to SEQ ID NO:1311 and one strand of said second dsRNA is substantially complementary to SEQ ID NO:1242 and wherein said first and second dsRNAs are less than 30 base pairs in length.
  • 31. An isolated cell comprising the vector of claim 30.
  • 32. The dsRNA of claim 2, comprising at least one 2′-O-Me ribonucleotide and at least one phosphorothioate.
  • 33. The dsRNA of claim 22, comprising at least one 2′-O-Me ribonucleotide and at least one phosphorothioate.
RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 60/787,762, filed Mar. 31, 2006, and U.S. Provisional Application No. 60/870,259, filed Dec. 15, 2006. Both prior applications are incorporated herein by reference in their entirety.

US Referenced Citations (35)
Number Name Date Kind
4426330 Sears Jan 1984 A
4534899 Sears Aug 1985 A
4837028 Allen Jun 1989 A
4868116 Morgan et al. Sep 1989 A
4980286 Morgan et al. Dec 1990 A
5013556 Woodle et al. May 1991 A
5139941 Muzyczka et al. Aug 1992 A
5213804 Martin et al. May 1993 A
5225212 Martin et al. Jul 1993 A
5252479 Srivastava Oct 1993 A
5264221 Tagawa et al. Nov 1993 A
5328470 Nabel et al. Jul 1994 A
5334711 Sproat et al. Aug 1994 A
5356633 Woodle et al. Oct 1994 A
5540935 Miyazaki et al. Jul 1996 A
5543152 Webb et al. Aug 1996 A
5556948 Tagawa et al. Sep 1996 A
5595760 Cherif-Cheikh Jan 1997 A
5627053 Usman et al. May 1997 A
5665710 Rahman et al. Sep 1997 A
5672659 Shalaby et al. Sep 1997 A
5705188 Junichi et al. Jan 1998 A
5716824 Beigelman et al. Feb 1998 A
5854038 Sullenger et al. Dec 1998 A
5902880 Thompson May 1999 A
6054299 Conrad Apr 2000 A
6146886 Thompson Nov 2000 A
6150092 Uchida et al. Nov 2000 A
6395713 Beigelman et al. May 2002 B1
20030203844 Delfani et al. Oct 2003 A1
20040018176 Tolentino et al. Jan 2004 A1
20040180847 Dobie et al. Sep 2004 A1
20040209832 McSwiggen et al. Oct 2004 A1
20050255487 Khvorova et al. Nov 2005 A1
20070031844 Khvorova et al. Feb 2007 A1
Foreign Referenced Citations (48)
Number Date Country
101 00 586 Apr 2002 DE
0 445 131 Oct 1989 EP
0 496 813 Oct 1990 EP
WO 8804924 Jul 1988 WO
WO 8902468 Mar 1989 WO
WO 8905345 Jun 1989 WO
WO 8907136 Aug 1989 WO
WO 9004384 May 1990 WO
WO 9105545 May 1991 WO
WO 9207065 Apr 1992 WO
WO 9207573 May 1992 WO
WO 9315187 May 1993 WO
WO 9323569 Nov 1993 WO
WO 9324641 Dec 1993 WO
WO 9402595 Feb 1994 WO
WO 9413788 Jun 1994 WO
WO 9420073 Sep 1994 WO
WO 9610390 Apr 1996 WO
WO 9610391 Apr 1996 WO
WO 9610392 Apr 1996 WO
WO 9640062 Dec 1996 WO
WO 9704787 Feb 1997 WO
WO 9713499 Apr 1997 WO
WO 97026270 Jul 1997 WO
WO 9730731 Aug 1997 WO
WO 9813526 Apr 1998 WO
WO 9932619 Jul 1999 WO
WO 9953050 Oct 1999 WO
WO 9961631 Dec 1999 WO
WO 0022113 Apr 2000 WO
WO 0022114 Apr 2000 WO
WO 0044895 Aug 2000 WO
WO 0053722 Sep 2000 WO
WO 03070910 Aug 2003 WO
WO 2003070918 Aug 2003 WO
WO 2004001193 Dec 2003 WO
WO 2004007070 Jan 2004 WO
WO 2004011822 Feb 2004 WO
WO 2004011829 Feb 2004 WO
WO 2004064737 Aug 2004 WO
WO 2004065601 Aug 2004 WO
WO 2004080406 Sep 2004 WO
WO 2004094345 Nov 2004 WO
WO 2004094595 Nov 2004 WO
WO 2004099410 Nov 2004 WO
WO 2005014782 Feb 2005 WO
WO 2005031002 Apr 2005 WO
WO 2007115168 Oct 2007 WO
Related Publications (1)
Number Date Country
20070281899 A1 Dec 2007 US
Provisional Applications (2)
Number Date Country
60787762 Mar 2006 US
60870259 Dec 2006 US