Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF EG5 AND VEGF GENES
Document Type and Number:
WIPO Patent Application WO/2009/111658
Kind Code:
A3
Abstract:
This invention relates to compositions containing double-stranded ribonucleic acid (dsRNA) in a SNALP formulation, and methods of using the compositions to inhibit the expression of the Eg5 and Vascular Endothelial Growth Factor (VEGF), and methods of using the compositions to treat pathological processes mediated by Eg5 and VEGF expression, such as cancer.

Inventors:
BUMCROT DAVID (US)
Application Number:
PCT/US2009/036223
Publication Date:
October 29, 2009
Filing Date:
March 05, 2009
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ALNYLAM PHARMACEUTICALS INC (US)
BUMCROT DAVID (US)
International Classes:
A61K31/713; C07H21/00; C12N15/113
Domestic Patent References:
WO2007115168A22007-10-11
WO2007012191A12007-02-01
WO2004065601A22004-08-05
WO2005089224A22005-09-29
Other References:
ZIMMERMANN TRACY S; ET AL: "RNAi-mediated gene silencing in non-human primates", NATURE, vol. 441, no. 7089, 4 May 2006 (2006-05-04), pages 111 - 114, XP002412249, ISSN: 0028-0836
TAKIMOTO CHRIS H; AWADA AHMAD: "Safety and anti-tumor activity of sorafenib (Nexavar((R))) in combination with other anti-cancer agents: a review of clinical trials", CANCER CHEMOTHERAPY AND PHARMACOLOGY, vol. 61, no. 4, April 2008 (2008-04-01), pages 535 - 548, XP002543476, ISSN: 0344-5704
ZHU ANDREW X: "Development of sorafenib and other molecularly targeted agents in hepatocellular carcinoma", CANCER, vol. 112, no. 2, January 2008 (2008-01-01), pages 250 - 259, XP002543477, ISSN: 0008-543X
Attorney, Agent or Firm:
SHUSTER, Michael, J. et al. (Silicon Valley Center801 California Stree, Mountain View CA, US)
Download PDF:
Claims:

CLAIMS

We claim:

1. A composition comprising a first double-stranded ribonucleic acid (dsRNA) for inhibiting the expression of a human kinesin family member 11 (Eg5/KSP) gene in a cell and a second dsRNA for inhibiting expression of a human VEGF in a cell, wherein: both said first and said second dsRNA are formulated in a stable nucleic acid lipid particle (SNALP); said first dsRNA consists of a first sense strand and a first antisense strand, and said first sense strand comprises a first sequence and said first antisense strand comprises a second sequence complementary to at least 15 contiguous nucleotides of SEQ ID NO: 1311 ( 5 ' - UCGAGAAUCUAAACUAACU- 3 ' ) , wherein said first sequence is complementary to said second sequence and wherein said first dsRNA is between 15 and 30 base pairs in length; and said second dsRNA consists of a second sense strand and a second antisense strand, said second sense strand comprising a third sequence and said second antisense strand comprising a fourth sequence complementary to at least 15 contiguous nucleotides of SEQ ID NO: 1538 (5'-GCACAUAGGAGAGAUGAGCUU- S ' ) , wherein said third sequence is complementary to said fourth sequence and wherein each strand is between 15 and 30 base pairs in length.

2. The composition of claim 1, wherein the first antisense strand comprises a second sequence complementary to SEQ ID NO: 1311 ( 5 ' -UCGAGAAUCUAAACUAACU- 3 ' ) and the second antisense strand comprises a fourth sequence complementary to SEQ ID NO: 1538

(5'-GCACAUAGGAGAGAUGAGCUU- S ' ) . .

3. The composition of claim 1, wherein the first dsRNA consists of a sense strand consisting of SEQ ID NO: 1534 (5 '-UCGAGAAUCUAAACUAACUTT- S ' ) and an antisense strand consisting of SEQ ID NO: 1535 (5 '-AGUUAGUUUAGAUUCUCGATT - S ' ) and the second dsRNA consists of a sense strand consisting of SEQ ID NO: 1536 (5'-

GCACAUAGGAGAGAUGAGCUU- 3 ' ) , and an antisense strand consisting of SEQ ID NO: 1537 (5'-AAGCUCAUCUCUCCUAUGUGCUG- S ' ) .

4. The composition of claim 3, 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": the first dsRNA consists of a sense strand consisting of SEQ ID NO: 1240 (S'-ucGAGAAucuAAAcuAAcuTsT-S')

and an antisense strand consisting of

SEQ ID NO: 1241 (S'-AGUuAGUUuAGAUUCUCGATsT); the second dsRNA consists of a sense strand consisting of

SEQ ID NO: 1242 (S'-GcAcAuAGGAGAGAuGAGCUsU-S') and an antisense strand consisting of

SEQ ID NO: 1243 (S'-AAGCUcAUCUCUCCuAuGuGCusG-S').

5. The composition of claims 1, 2, or 3, wherein said first and second dsRNA comprises at least one modified nucleotide.

6. The composition of claim 5, 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.

7. The composition of claim 5, 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-modifϊed nucleotide, 2'-alkyl-modifϊed nucleotide, morpholino nucleotide, a phosphoramidate, and a non-natural base comprising nucleotide.

8. The composition of claims 1, 2, and 3, wherein said first and second dsRNA each comprise at least one 2'-O-methyl modified ribonucleotide and at least one nucleotide comprising a 5'-phosphorothioate group.

9. The composition of claims 1-3 and 5-8, wherein each strand of each dsRNA is 19-23 bases in length.

10. The composition of claims 1-3 and 5-8, wherein each strand of each dsRNA is 21-23 bases in length.

11. The composition of claims 1-3 and 5-8, wherein each strand of the first dsRNA is 21 bases in length and the sense strand of the second dsRNA is 21 bases in length and the antisense strand of the second dsRNA is 23 bases in length.

12. The composition of claims 1-11, wherein the first and second dsRNA are present in an equimolar ratio.

13. The composition of claims 1-12, wherein said SNALP comprises DLinDMA, cholesterol, DPPC, and PEG2000-C-DMA.

14. The composition of claims 1-13, comprising the components in the proportions listed in Table 17.

15. The composition of claims 1-14, wherein said composition, upon contact with a cell expressing Eg5, inhibits expression of Eg5 by at least 40, 50, 60, 70, 80, or by at least 90%.

16. The composition of claims 1-15, wherein said composition, upon contact with a cell expressing VEGF, inhibits expression of VEGF by at least 40, 50, 60, 70, 80, or by at least 90%.

17. The composition of claims 1-16, wherein administration of said composition to a cell decreases expression of both Eg5 and VEGF in said cell.

18. The composition of claims 1-17, wherein the composition is administered in a nM concentration.

19. The composition of claims 1-18, wherein administration of said composition to a cell increases mono-aster formation in the cell.

20. The composition of claims 1-19, wherein administration of said composition to a mammal results in at least one effect selected from the group consisting of prevention of tumor growth, reduction in tumor growth, or prolonged survival in said mammal.

21. The composition of claims 1-20, wherein said effect is measured using at least one assay selected from the group consisting of determination of body weight, determination of organ weight, visual inspection, mRNA analysis, serum AFP analysis and survival monitoring.

22. The composition of claims 1-21, further comprising Sorafenib.

23. The composition of any of the above claims, wherein the first dsRNA contains two overhangs and the second dsRNA contains an overhang at the 3' of the antisense and a blunt end at the 5' end of the antisense strand.

24. A method for inhibiting the expression of Eg5/KSP and VEGF in a cell comprising administering any of the compositions of claims 1-22 to the cell.

25. A method for preventing tumor growth, reducing tumor growth, or prolonging survival in a mammal in need of treatment for cancer comprising administering the composition of claims 1-22 to said mammal.

26. The method of claim 25, wherein said mammal has liver cancer.

27. The method of claim 25, wherein said mammal is a human with liver cancer.

28. The method of claim 24 or 25, further comprising administering Sorafenib.

Description:

COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5

AND VEGF GENES

Related Applications

This application claims the benefit of U.S. Provisional Application No. 61/034,019, filed March 5, 2008, and U.S. Provisional Application No. 61/083,367, filed July 24, 2008, and U.S. Provisional Application No. 61/086,381, filed August 5, 2008, and U.S. Provisional Application No. 61/112,079, filed November 6, 2008, and U.S. Provisional Application No. 61/150,664, filed February 6, 2009, which are hereby incorporated in their entirety by reference.

Field of the Invention

This invention relates to compositions containing double-stranded ribonucleic acid (dsRNA), and their use in mediating RNA interference to inhibit the expression of a combination of genes, e.g., the Eg5 and Vascular Endothelial Growth Factor (VEGF) genes formulated in SNALP, and the use of the compositions to treat pathological processes mediated by Eg5 and VEGF expression, such as cancer.

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 induce 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 have a role. Hence, there is also a need for agents 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. M., 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 an 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.

Summary of the Invention

Disclosed are compositions having two double-stranded ribonucleic acids (dsRNA) for inhibiting the expression of a human kinesin family member 11 (Eg5/KSP) and a human VEGF gene in a cell. The dsRNAs are formulated in a stable nucleic acid lipid particle (SNALP). Also disclosed are method for using the composition to decrease expression of Eg5/KSP and/or VEGF in a cell, and method of treatment of a disease, e.g., liver cancer, using the compositions of the invention.

Accordingly, disclosed herein is a composition having a first double-stranded ribonucleic acid (dsRNA) for inhibiting the expression of a human kinesin family member 11 (Eg5/KSP) gene in a cell and a second dsRNA for inhibiting expression of a human VEGF in a cell, wherein both said first and said second dsRNA are formulated in a stable nucleic acid lipid particle (SNALP); said first dsRNA consists of a first sense strand and a first antisense strand, and said first sense strand has a first sequence and said first antisense strand has a second sequence complementary to at least 15 contiguous nucleotides of SEQ ID NO: 1311

(5'-UCGAGAAUCUAAACUAACU-S '), wherein said first sequence is complementary to said second sequence and wherein said first dsRNA is between 15 and 30 base pairs in length; and said second dsRNA consists of a second sense strand and a second antisense strand, said second sense strand having a third sequence and said second antisense strand having a fourth sequence complementary to at least 15 contiguous nucleotides of SEQ ID NO: 1538 (5 '-GCACAUAGGAGAGAUGAGCUU-S '), wherein said third sequence is complementary to said fourth sequence and wherein each strand is between 15 and 30 base pairs in length.

In some embodiments, the first antisense strand has a second sequence complementary to SEQ ID NO: 1311 (5'-UCGAGAAUCUAAACUAACU-S ') and the second antisense strand has a fourth sequence complementary to SEQ ID NO: 1538 (5'- GCACAUAGGAGAGAUGAGCUU-3'). In other embodiments, the first dsRNA consists of a sense strand consisting of SEQ ID NO: 1534 (5'-UCGAGAAUCUAAACUAACUTT-S ') and an antisense strand consisting of SEQ ID NO: 1535 (5'-

AGUUAGUUUAGAUUCUCGATT-3') and the second dsRNA consists of a sense strand consisting of SEQ ID NO: 1536 (5'-GCACAUAGGAGAGAUGAGCUU-S '), and an antisense strand consisting of SEQ ID NO: 1537 (5'-AAGCUCAUCUCUCCUAUGUGCUG- 3'). In further embodiments, 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": the first dsRNA consists of a sense strand consisting of SEQ ID NO: 1240 (S'-ucGAGAAucuAAAcuAAcuTsT-S'), and an antisense strand consisting of SEQ ID NO: 1241 (S'-AGUuAGUUuAGAUUCUCGATsT); the second dsRNA consists of a sense strand consisting of SEQ ID NO: 1242 (S'-GcAcAuAGGAGAGAuGAGCUsU-S') and an antisense strand consisting of SEQ ID NO: 1243 (5'- AAGCUcAUCUCUCCuAuGuGCusG-3 ').

In some embodiments, the first dsRNA contains two overhangs and the second dsRNA contains an overhang at the 3' of the antisense and a blunt end at the 5' end of the antisense strand.

The first and second dsRNA can have at least one modified nucleotide. For example, each dsRNA can have at least one modified nucleotide chosen from the group of: a 2'-O- methyl modified nucleotide, a nucleotide having a 5'-phosphorothioate group, and a terminal nucleotide linked to a cholesteryl derivative or dodecanoic acid bisdecylamide group. The modified nucleotide can be chosen from the group of: a 2'-deoxy-2'-fluoro modified nucleotide, a 2'-deoxy-modified nucleotide, a locked nucleotide, an abasic nucleotide, T-

amino-modified nucleotide, 2'-alkyl-modified nucleotide, morpholino nucleotide, a phosphoramidate, and a non-natural base having nucleotide. In some embodiments, the first and second dsRNA each comprise at least one 2'-O-methyl modified ribonucleotide and at least one nucleotide having a 5'-phosphorothioate group.

Each strand of each dsRNA can be, e.g., 19-23 bases in length, or, alternatively 21-23 bases in length. In one embodiment, each strand of the first dsRNA is 21 bases in length and the sense strand of the second dsRNA is 21 bases in length and the antisense strand of the second dsRNA is 23 bases in length.

In some embodiments, the first and second dsRNA are present in an equimolar ratio.

As described herein, the dsRNAs are formulated as SNALPS. In some embodiments, the SNALP formulation includes DLinDMA, cholesterol, DPPC, and PEG2000-C-DMA. For example, the SNALP can have the components in the proportions listed in Table 17.

The composition of the invention can be used to reduce expression of Eg5 and/or VAGF. In some embodiments, the composition of the invention, upon contact with a cell expressing Eg5, inhibits expression of Eg5 by at least 40, 50, 60, 70, 80, or by at least 90%. In other embodiments, the composition of the invention, upon contact with a cell expressing VEGF, inhibits expression of VEGF by at least 40, 50, 60, 70, 80, or by at least 90%. Administration of the composition to a cell can expression of both Eg5 and VEGF in said cell. The composition of claims 1-17, wherein the composition is administered in a nM concentration.

Administration of the composition of the invention to a cell can result in, e.g., an increase in mono-aster formation in the cell. Administration of the composition to a mammal can result in at least one effect selected from the group consisting of prevention of tumor growth, reduction in tumor growth, or prolonged survival in said mammal. The effect can be measured using at least one assay selected from the group consisting of determination of body weight, determination of organ weight, visual inspection, mRNA analysis, serum AFP analysis and survival monitoring. Included are compositions with these effect when administered in a nM concentration.

In a further embodiment the composition of the invention includes Sorafenib.

Also included in the invention are methods of suing the compositions of the invention. In one embodiment is are methods for inhibiting the expression of Eg5/KSP and VEGF in a cell by administering any of the compositions of the invention to the cell. Other embodiments are methods for preventing tumor growth, reducing tumor growth, or prolonging survival in a mammal in need of treatment for cancer by administering the

composition to said mammal. In some embodiments the mammal has liver cancer, e.g., the mammal is a human with liver cancer. The method can include a further step of administering Sorafenib.

Brief Description of the Figures

FIG. 1 is a graph showing liver weights as percentage of body weight following administration of SNALP-siRNAs in a Hep3B mouse model.

FIGs. 2A-2D are graphs showing the effects of SNALP-siRNAs on body weight in a Hep3B mouse model.

FIG. 3 is a graph showing the effects of SNALP-siRNAs on body weight in a Hep3B mouse model.

FIG. 4 is a graph showing the body weight in untreated control animals.

FIG. 5 is a graph showing the effects of control luciferase- SNALP siRNAs on body weight in a Hep3B mouse model.

FIG. 6 is a graph showing the effects of VSP-SNALP siRNAs on body weight in a Hep3B mouse model.

FIG. 7A is a graph showing the effects of SNALP-siRNAs on human GAPDH levels normalized to mouse GAPDH levels in a Hep3B mouse model.

FIG. 7B is a graph showing the effects of SNALP-siRNAs on serum AFP levels as measured by serum ELISA in a Hep3B mouse model.

FIG. 8 is a graph showing the effects of SNALP-siRNAs on human GAPDH levels normalized to mouse GAPDH levels in a Hep3B mouse model.

FIG. 9 is a graph showing the effects of SNALP-siRNAs on human KSP levels normalized to human GAPDH levels in a Hep3B mouse model.

FIG. 10 is a graph showing the effects of SNALP-siRNAs on human VEGF levels normalized to human GAPDH levels in a Hep3B mouse model.

FIG. 1 IA is a graph showing the effects of SNALP-siRNAs on mouse VEGF levels normalized to human GAPDH levels in a Hep3B mouse model.

FIG. 1 IB is a set of graphs showing the effects of SNALP-siRNAs on human GAPDH levels and serum AFP levels in a Hep3B mouse model.

FIGs. 12A-12C are graphs showing the effects of SNALP-siRNAs on tumor KSP, VEGF and GAPDH levels in a Hep3B mouse model.

FIG. 13 A and FIG. 13B are graphs showing the effects of SNALP-siRNAs on survival in mice with hepatic tumors. Treatment was started at 18 days (FIG. 13A) and 26 days (FIG. 13B) after tumor cell seeding.

FIG. 14 is a graph showing the effects of SNALP-siRNAs on serum alpha fetoprotein (AFP) levels.

FIG. 15A and 15B are images of H&E stained sections in tumor bearing animals (three weeks after Hep3B cell implantation) were administered 2 mg/kg SNALP-VSP (A) or 2 mg/kg SNALP-Luc (B). Twenty four hours later, tumor bearing liver lobes were processed for histological analysis. Arrows indicate mono asters.

FIG. 16 is a flow diagram illustrating the manufacturing process of ALN-VSPDSOl.

FIG. 17 is a cryo-transmission electron microscope (cryo-TEM) image of ALN- VSP02.

FIG. 18 is a flow diagram illustrating the manufacturing process of ALN-VSP02.

FIG. 19 is a graph illustrating the effects on survival of administration SNALP formulated siRNA and Sorafenib.

Detailed Description of the Invention

The invention provides compositions and methods for inhibiting the expression of the Eg5 gene and VEGF gene in a cell or mammal using the dsRNAs. The dsRNAs are preferably packaged in a stable nucleic acid particle (SNALP). The invention also provides compositions and methods for treating pathological conditions and diseases, such as liver cancer, in a mammal caused by the expression of the Eg5 gene and VEGF genes. The dsRNA directs the sequence-specific degradation of mRNA through a process known as RNA interference (RNAi).

The following detailed description discloses how to make and use the compositions containing dsRNAs to inhibit the expression of the Eg5 gene and VEGF genes, respectively, as well as compositions and methods for treating diseases and disorders caused by the expression of these genes, such as cancer. The pharmaceutical compositions featured in the invention include 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. The compositions featured in the invention also include a dsRNA having an antisense strand having 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 VEGF gene.

Accordingly, certain aspects of the invention provide pharmaceutical compositions containing the Eg5 and VEGF dsRNAs and a pharmaceutically acceptable carrier, methods of using the compositions to inhibit expression of the Eg5 gene and the VEGF gene respectively, and methods of using the pharmaceutical compositions to treat diseases caused by expression of the Eg5 and VEGF genes.

L 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. "T" and "dT" are used interchangeably herein and refer to a deoxyribonucleotide wherein the nucleobase is thymine, e.g., deoxyribothymine. 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. In another example, adenine and cytosine anywhere in the oligonucleotide can be replaced with guanine and uracil, respectively to form G-U Wobble base pairing with the target mRNA. Sequences comprising such replacement moieties are embodiments of the invention.

As used herein, "Eg5" refers to the human kinesin family member 11, which is also known as KIFl 1, Eg5, HKSP, KSP, KNSLl or TRIP5. Eg5 sequence can be found as NCBI GeneID:3832, HGNC ID: HGNC:6388 and RefSeq ID number:NM_004523. The terms "Eg5" and "KSP" and "Eg5/KSP are used interchangeably

As used herein, 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 protein 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. VEGF 145 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 FIt-I (VEGFR-I) 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, VEGF 145 induces angiogenesis and like VEGF 189 (but unlike VEGF 165) VEGF 145 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 US Ser. No. 11/078,073 and 11/340,080, which are hereby incorporated by reference in their entirety.

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/KSP and/or VEGF gene, including mRNA that is a product of RNA processing of a primary transcription product.

As used herein, the term "strand comprising a sequence" refers to an oligonucleotide comprising a chain of nucleotides that is described by the sequence referred to using the standard nucleotide nomenclature.

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 0 C or 70 0 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. Such non- Watson-Crick base pairs includes, but not limited to, G:U Wobble or Hoogstein base pairing.

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/KSP and/or VEGF) including a 5' UTR, an open reading frame (ORF), or a 3' UTR. 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 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. In general, the majority of nucleotides of each strand are ribonucleotides, but as described in detail herein, each or both strands can also include at least one non-ribonucleotide, e.g., a deoxyribonucleotide and/or a modified nucleotide. In addition, as used in this specification, "dsRNA" may include chemical modifications to ribonucleotides, including substantial modifications at multiple nucleotides and including all types of modifications disclosed herein or known in the art. Any such modifications, as used in an siRNA type molecule, are encompassed by "dsRNA" for the purposes of this specification and claims.

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. In some embodiments the dsRNA can have a nucleotide overhang at one end of the duplex and a blunt end at the other end.

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 may be in the internal or terminal regions of the molecule. Generally the most tolerated

mismatches are in the terminal 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.

As used herein, the term "SNALP" refers to a stable nucleic acid-lipid particle. A SNALP represents a vesicle of lipids coating a reduced aqueous interior comprising a nucleic acid such as an iRNA agent or a plasmid from which an iRNA agent is transcribed.

"Introducing into a cell", when referring to a dsRNA, means facilitating uptake or absorption into the cell, as is understood by those skilled in the art. Absorption or uptake of dsRNA can occur through unaided diffusive or active cellular processes, or by auxiliary agents or devices. The meaning of this term is not limited to cells in vitro; a dsRNA may also be "introduced into a cell", wherein the 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 "down-regulate the expression of," "suppress the expression of and the like in as far as they refer to the Eg5 and/or VEGF 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 Eg5 mRNA and/or VEGF mRNA which may be isolated from a first cell or group of cells in which the Eg5 and/or VEGF gene is transcribed and which has or have been treated such that the expression of the Eg5 and/or VEGF 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) • IuU /o

(mRNA in control cells)

Alternatively, the degree of inhibition may be given in terms of a reduction of a parameter that is functionally linked to Eg5 and/or VEGF gene expression, e.g. the amount of protein encoded by the Eg5 and/or VEGF gene which is produced by a cell, or the number of cells displaying a certain phenotype, e.g. apoptosis. In principle, target gene silencing can 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 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% by administration of the double-stranded oligonucleotide of the invention. In some embodiments, the Eg5 and/or VEGF gene is suppressed by at least about 60%, 70%, or 80% by administration of the double-stranded oligonucleotide of the invention. In other embodiments, the Eg5 and/or VEGF gene is suppressed by at least about 85%, 90%, or 95% by administration of the double-stranded oligonucleotide of the invention. The Tables and Example below provides values for inhibition of expression using various Eg5 and/or VEGF dsRNA molecules at various concentrations.

As used herein in the context of Eg5 expression (or VEGF expression), the terms "treat", "treatment", and the like, refer to relief from or alleviation of pathological processes mediated by Eg5 and/or VEGF 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 and/or VEGF 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 and/or VEGF expression or an overt symptom of pathological processes mediated by Eg5 and/or 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 and/or VEGF expression, the patient's history and age, the stage of pathological processes mediated by Eg5 and/or VEGF expression, and the administration of other anti-pathological processes mediated by Eg5 and/or VEGF 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. As described in more detail below, 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)

As described in more detail below, the invention provides double-stranded ribonucleic acid (dsRNA) molecules for inhibiting the expression of the Eg5 and/or VEGF gene 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 and/or VEGF 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 and/or VEGF gene, inhibits the expression of said Eg5 and/or VEGF gene.

The dsRNA can be synthesized by standard 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.

The dsRNA comprises two 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 and/or VEGF 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 is between 15 and 30, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 base pairs in length. In other embodiments the duplex structure is 25-30 base pairs in length.

In one embodiment the duplex is 19 base pairs in length. In another embodiment the duplex is 21 base pairs in length. When two different siRNAs are used in combination, the duplex lengths can be identical or can differ. For example, a composition can include a first dsRNA targeted to Eg5 with a duplex length of 19 base pairs and a second dsRNA targeted to VEGF with a duplex length of 21 base pairs.

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. In other embodiments the region of complementarity is 25-30 nucleotides in length.

In one embodiment the region of complementarity is 19 nucleotides in length. In another embodiment the region of complementarity is 21 nucleotides in length. When two different siRNAs are used in combination, the region of complementarity can be identical or can differ. For example, a composition can include a first dsRNA targeted to Eg5 with a region of complementarity of 19 nucleotides and a second dsRNA targeted to VEGF with a region of complementarity of 21 nucleotides.

Each strand of the dsRNA of invention is generally between 15 and 30, or between 18 and 25, or 18, 19, 20, 21, 22, 23, or 24 nucleotides in length. In other embodiments, each is strand is 25-30 base pairs in length. Each strand of the duplex can be the same length or of different lengths. When two different siRNAs are used in combination, the lengths of each strand of each siRNA can be identical or can differ. For example, a composition can include a dsRNA targeted to Eg5 with a sense strand of 21 nucleotides and an antisense strand of 21 nucleotides, and a second dsRNA targeted to VEGF with a sense strand of 21 nucleotides and an antisense strand of 23 nucleotides.

The dsRNA of the invention can include one or more single-stranded overhang(s) of one or more nucleotides. 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. In another embodiment,

the antisense strand of the dsRNA has 1-10 nucleotides overhangs each at the 3' end and the 5' end over the sense strand. In further embodiments, the sense strand of the dsRNA has 1- 10 nucleotides overhangs each at the 3' end and the 5' end over the antisense strand.

A dsRNAs having at least one nucleotide overhang can have unexpectedly superior inhibitory properties than the blunt-ended counterpart. In some embodiments the presence of only one nucleotide overhang strengthens the interference activity of the dsRNA, without affecting its overall stability. A 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 can also have a blunt end, generally located at the 5 '-end of the antisense strand. Such dsRNAs can have improved stability and inhibitory activity, thus allowing administration at low dosages, i.e., 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.

As described in more detail herein, the composition of the invention includes a first dsRNA targeting Eg5 and a second dsRNA targeting VEGF. The first and second dsRNA can have the same overhang architecture, e.g., number of nucleotide overhangs on each strand, or each dsRNA can have a different architecture. In one embodiment, the first dsRNA targeting Eg5 includes a 2 nucleotide overhang at the 3 ' end of each strand and the second dsRNA targeting VEGF includes a 2 nucleotide overhang on the 3' end of the antisense strand and a blunt end at the 5' end of the antisense strand (e.g., the 3' end of the sense strand).

In one embodiment, the Eg5 gene targeted by the dsRNA of the invention is the human Eg5 gene. In one embodiment, the antisense strand of the dsRNA targeting Eg5 comprises at least 15 contiguous nucleotides of one of the antisense sequences of Table 1-3. In specific embodiments, the first sequence of the dsRNA is selected from one of the sense strands 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 some embodiments the dsRNA targeted to Eg5 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

In embodiments using a second dsRNA targeting VEGF, such agents are exemplified in the Examples, Tables 4a and 4b, and in co-pending US Serial Nos: 11/078,073 and 11/340,080, herein incorporated by reference. In one embodiment the dsRNA targeting VEGF has an antisense strand complementary to at least 15 contiguous nucleotides of the VEGF target sequences described in Table 4a. In other embodiments, the dsRNA targeting VEGF comprises one of the antisense sequences of Table 4b, or one of the sense sequences of Table 4b, or comprises one of the duplexes (sense and antisense strands) of Table 4b.

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 ah, 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. Additional dsRNA targeting VEGF can be designed in a similar matter using the sequences disclosed in Tables 4a and 4b, the Examples and co-pending US Serial Nos: 11/078,073 and 11/340,080, herein incorporated by reference.

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, e.g., dsRNA, 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. Additional RNAi agents, e.g., dsRNA, targeting VEGF can be designed in a similar matter using the sequences disclosed in Tables 4a and 4b, the Examples and co-pending US Serial Nos: 11/078,073 and 11/340,080, herein incorporated by reference.

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.

Modifications

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, Inc., New York, NY, 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 is 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 cycloalkyl 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, an 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 ah, 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 2 -NH- CH 2 -, — CH 2 -- N(CH 3 )- O— CH 2 -- [known as a methylene (methylimino) or MMI backbone], - -CH 2 -O-N(CH 3 )-CH 2 -, -CH 2 -N(CH 3 )-N(CH 3 )-CH 2 - and -N(CH 3 )-CH 2 -CH 2 - [wherein the native phosphodiester backbone is represented as -0-P-O-CH 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 morpholino 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-0-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted Ci to C 10 alkyl or C 2 to C 10 alkenyl and alkynyl. Particularly preferred are O[(CH 2 ) n O] m CH 3 , O(CH 2 ) n OCH 3 , O(CH 2 ) n NH 2 , O(CH 2 ) n CH 3 , O(CH 2 ) n ONH 2 , and O(CH 2 ) n ON[(CH 2 ) n CH 3 )] 2 , where n and m are from 1 to about 10. Other preferred dsRNAs comprise one of the following at the 2' position: Ci to C 10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH 3 , OCN, Cl, Br, CN, CF 3 , OCF 3 , SOCH 3 , SO 2 CH 3 , ONO 2 , NO 2 , N 3 , NH 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'-0— CH 2 CH 2 OCH 3 , also known as 2'-O-(2-methoxyethyl) or 2'-MOE) (Martin et ah, HeIv. Chim.

Acta, 1995, 78, 486-504) i.e., an alkoxy-alkoxy group. A further preferred modification includes 2'-dimethylaminooxyethoxy, i.e., a O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as T- DMAOE, as described in examples herein below, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-O-dimethylaminoethoxyethyl or 2'-DMAEOE), i.e., 2'-0-CH 2 -O- CH 2 - N(CH 2 ) 2 , also described in examples herein below.

Other preferred modifications include 2'-methoxy (2'-OCH 3 ), 2'-aminopropoxy (T- OCH 2 CH 2 CH 2 NH 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 nucleotide or in 2'-5' linked dsRNAs and the 5' position of 5' terminal nucleotide. DsRNAs may also have sugar mimetics such as cyclobutyl 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 purine 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-halouracil 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 cytosine's, 7-methylguanine and 7- methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-daazaadenine and 3- deazaguanine and 3-deazaadenine. 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 ah, Angewandte 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- propynylcytosine. 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- methoxy ethyl 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.

Conjugates

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 ah, Proc. Natl. Acid. Sci. USA, 199, 86, 6553-6556), cholic acid (Manoharan et al, Biorg. Med. Chem. Let, 1994 4 1053-1060), a thioether, e.g., beryl-S-tritylthiol (Manoharan et al, Ann. N.Y. Acad. Sci., 1992, 660, 306- 309; Manoharan et ah, Biorg. Med. Chem. Let, 1993, 3, 2765-2770), a thiocholesterol (Oberhauser et ah, Nucl. Acids Res., 1992, 20, 533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et ah, EMBO J, 1991, 10, 1111-1118; Kabanov et ah, FEBS Lett., 1990, 259, 327-330; Svinarchuk et ah, Biochimie, 1993, 75, 49-54), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethyl-ammonium 1,2-di-O-hexadecyl-rac- glycero-3-Hphosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654; Shea et ah, Nucl. Acids Res., 1990, 18, 3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et ah, Nucleosides & Nucleotides, 1995, 14, 969-973), or adamantane acetic acid (Manoharan et ah, Tetrahedron Lett., 1995, 36, 3651-3654), a palmityl moiety (Mishra et ah, 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; 4,667,025; 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; 5,245,022; 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 enzymes 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:DNA duplex. 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 deoxy dsRNAs 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 ah, Proc. Natl. Acad. Sci. USA, 1989, 86:6553), cholic acid (Manoharan et ah, Bioorg. Med. Chem. Lett., 1994, 4: 1053), a

thioether, e.g., hexyl-S-tritylthiol (Manoharan et ah, Ann. N.Y. Acad. Sci., 1992, 660:306; Manoharan et ah, Bioorg. Med. Chem. Let., 1993, 3:2765), a thiocholesterol (Oberhauser et ah, Nucl. Acids Res., 1992, 20:533), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et ah, EMBO J., 1991, 10: 111; Kabanov eϊ α/., FEBS Lett., 1990, 259:327; Svinarchuk et ah, Biochimie, 1993, 75:49), a phospholipid, e.g., di-hexadecyl-rac- glycerol or triethylammonium l^-di-O-hexadecyl-rac-glycero-S-H-phosphonate (Manoharan et ah, Tetrahedron Lett., 1995, 36:3651; Shea et ah, Nucl. Acids Res., 1990, 18:3777), a polyamine or a polyethylene glycol chain (Manoharan et ah, Nucleosides & Nucleotides, 1995, 14:969), or adamantane acetic acid (Manoharan et ah, Tetrahedron Lett., 1995, 36:3651), a palmityl moiety (Mishra et ah, Biochim. Biophys. Acta, 1995, 1264:229), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et ah, J. Pharmacol. 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.

In some cases, a ligand can be multifunctional and/or a dsRNA can be conjugated to more than one ligand. For example, the dsRNA can be conjugated to one ligand for improved uptake and to a second ligand for improved release.

Vector encoded RNAi agents

In another aspect of the invention, Eg5 and VEGF specific dsRNA molecules that are expressed from transcription units inserted into DNA or RNA vectors (see, e.g., Couture, A, et ah, TIG. (1996), 12:5-10; Skillern, A., et ah, International PCT Publication No. WO 00/22113, Conrad, International PCT Publication No. WO 00/22114, and Conrad, US 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 ah, 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 a 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 recombinant 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 Muzyczka, 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 variety of genes into many different cell types, including epithelial cells, in vitro and/or in vivo (see, e.g., Eglitis, et al, Science (1985) 230: 1395-1398; Danos and Mulligan, Proc. Natl. Acad. ScL USA (1998) 85:6460-6464; Wilson et al, 1988, Proc. Natl. Acad. Sci. USA 85:3014-3018; Armentano et al, 1990, Proc. Natl. Acad. Sci. USA 87:61416145; Huber et al, 1991, Proc. Natl. Acad. Sci. USA 88:8039-8043; Ferry et al, 1991, Proc. Natl. Acad. Sci. USA 88:8377-8381; Chowdhury et al, 1991, Science 254: 1802- 1805; van Beusechem. et al, 1992, Proc. Natl. 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. Immunol. 150:4104-4115; U.S. Patent No. 4,868,116; U.S. Patent No. 4,980,286; PCT Application WO 89/07136; PCT Application WO 89/02468; PCT Application WO 89/05345; 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. Infectious Disease, 166:769), and also have the advantage of not requiring mitotically active cells for infection.

Any viral vector capable of accepting the coding sequences for the dsRNA molecule(s) to 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); herpes 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, Mokola, 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 ε 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 having, for example, either the U6 or Hl 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.

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. CMV early promoter or actin promoter or Ul snRNA promoter) or generally RNA polymerase III promoter (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 ah, 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 ah, 1994, FASEB J. 8:20-24). Such inducible expression systems, suitable for the control of transgene expression in cells or in mammals include regulation by ecdysone, by estrogen, progesterone, tetracycline, chemical inducers of dimerization, and isopropyl-beta-Dl -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 VEGF gene) or multiple Eg5 genes (or VEGF genes) over a period of a week or more are also contemplated by the invention. Successful introduction of the vectors of the invention into host cells can be monitored using various known methods. For example, transient transfection. can be signaled with a reporter, such as a fluorescent marker, such as Green Fluorescent Protein (GFP). Stable transfection of ex vivo cells can be ensured using markers that provide the transfected cell with resistance to specific environmental factors (e.g., antibiotics and drugs), such as hygromycin B resistance.

The Eg5 specific dsRNA molecules and VEGF specific dsRNA molecules can also be inserted into vectors and used as gene therapy vectors for human patients. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Patent 5,328,470) or by stereotactic injection (see e.g., Chen et al. (1994) Proc. Natl. Acad. Sci. USA 91:3054-3057). The pharmaceutical preparation of the

gene therapy vector can include the gene therapy vector in an acceptable diluent, or can include 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.

Pharmaceutical compositions containing dsRNA

In one embodiment, the invention provides pharmaceutical compositions containing a dsRNA, as described herein, and a pharmaceutically acceptable carrier and methods of administering the same. The pharmaceutical composition containing the dsRNA is useful for treating a disease or disorder associated with the expression or activity of a Eg5/KSP and/or VEGF gene, such as pathological processes mediated by Eg5/KSP and/or VEGF expression, e.g., liver cancer. Such pharmaceutical compositions are formulated based on the mode of delivery.

Dosage

The pharmaceutical compositions featured herein are administered in dosages sufficient to inhibit expression of EG5/KSP and/or VEGF genes. In general, a suitable dose of dsRNA will be in the range of 0.01 to 200.0 milligrams per kilogram body weight of the recipient per day, generally in the range of 1 to 50 mg per kilogram body weight per day. For example, the dsRNA can be administered at 0.01 mg/kg, 0.05 mg/kg, 0.5 mg/kg, 1 mg/kg, 1.5 mg/kg, 2 mg/kg, 3 mg/kg, 5.0 mg/kg, 10 mg/kg, 20 mg/kg, 30 mg/kg, 40 mg/kg, or 50 mg/kg per single dose.

The pharmaceutical composition can be administered once daily, or the dsRNA may be administered as two, three, or more sub-doses at appropriate intervals throughout the day . The effect of a single dose on EG5/KSP AND/OR VEGF levels is long lasting, such that subsequent doses are administered at not more than 7 day intervals, or at not more than 1, 2, 3, or 4 week intervals.

In some embodiments the dsRNA is administered 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/KSP AND/OR VEGF expression. Such models are used for in vivo testing of dsRNA, as well as for determining a therapeutically effective dose. A suitable mouse model is, for example, a mouse containing a plasmid expressing human EG5/KSP AND/OR VEGF. Another suitable mouse model is a transgenic mouse carrying a transgene that expresses human EG5/KSP AND/OR VEGF.

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 ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit high therapeutic indices are preferred.

The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of compositions featured in 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 methods featured in 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, as discussed above, the dsRNAs featured in the invention can be administered in combination with other known agents effective in treatment of pathological processes mediated by target gene 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.

Administration

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, and subdermal, oral or parenteral, e.g., subcutaneous.

Typically, when treating a mammal with hyperlipidemia, the dsRNA molecules are administered systemically via parental means. Parenteral administration includes intravenous, intra-arterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intraparenchymal, intrathecal or intraventricular, administration. For example, dsRNAs, conjugated or unconjugate or formulated with or without liposomes, can be administered intravenously to a patient. For such, a dsRNA molecule can be formulated into compositions such as sterile and non-sterile aqueous solutions, non-aqueous solutions in common solvents such as alcohols, or solutions in liquid or solid oil bases. Such solutions also can contain buffers, diluents, and other suitable additives. For parenteral, intrathecal, or intraventricular administration, a dsRNA molecule can be formulated into compositions such as sterile aqueous solutions, which also can contain buffers, diluents, and other suitable additives (e.g., penetration enhancers, carrier compounds, and other pharmaceutically acceptable carriers). Formulations are described in more detail herein.

The dsRNA can be delivered in a manner to target a particular tissue, such as the liver (e.g., the hepatocytes of the liver).

Formulations

The pharmaceutical formulations of the present invention, which may conveniently 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 then, 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.

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. In one aspect are formulations that target the liver when treating hepatic disorders such as hyperlipidemia.

In addition, dsRNA that target the EG5/KSP AND/OR VEGFgene can be formulated into compositions containing the dsRNA admixed, encapsulated, conjugated, or otherwise associated with other molecules, molecular structures, or mixtures of nucleic acids. For example, a composition containing one or more dsRNA agents that target the Eg5/KSP and/or VEGFgene can contain other therapeutic agents such as other cancer therapeutics or one or more dsRNA compounds that target non-EG5/KSP AND/OR VEGFgenes.

Oral, parenteral, topical, and biologic formulations

Compositions and formulations for oral administration include powders or granules, microparticulates, nanoparticulates, suspensions 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. In some embodiments, oral formulations are those in which dsRNAs featured in the invention are administered in conjunction with one or more penetration enhancers surfactants and chelators. Suitable surfactants include fatty acids and/or esters or salts thereof, bile acids and/or salts thereof. Suitable 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-dihydro-fusidate and sodium

glycodihydrofusidate. Suitable 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, glyceryl 1-monocaprate, 1- dodecylazacycloheptan-2-one, an acylcarnitine, an acylcholine, or a monoglyceride, a diglyceride or a pharmaceutically acceptable salt thereof (e.g., sodium). In some embodiments, combinations of penetration enhancers are used, for example, fatty acids/salts in combination with bile acids/salts. One exemplary 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 featured in 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, polyalkylcyanoacrylates; cationized gelatins, albumins, starches, acrylates, polyethyleneglycols (PEG) and starches; polyalkylcyanoacrylates; DEAE-derivatized polyimines, pollulans, celluloses and starches. Suitable complexing agents include chitosan, N-trimethylchitosan, poly-L-lysine, polyhistidine, polyornithine, polyspermines, protamine, polyvinylpyridine, polythiodiethylaminomethylethylene P(TDAE), polyaminostyrene (e.g., p-amino), poly(methylcyanoacrylate), poly(ethylcyanoacrylate), poly(butylcyanoacrylate), poly(isobutylcyanoacrylate), poly(isohexylcynaoacrylate), 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. Patent 6,887,906, U.S. patent publication. No. 20030027780, and U.S. Patent No. 6,747,014, each of which is incorporated herein by reference.

Compositions and formulations for parenteral, intraparenchymal (into the brain), 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 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. Suitable topical formulations include those in which the dsRNAs featured in the invention are in admixture with a topical delivery agent

such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents and surfactants. Suitable 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 featured in 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. Suitable fatty acids and esters include but are not limited to arachidonic acid, oleic acid, eicosanoic acid, lauric acid, caprylic acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein, dilaurin, glyceryl 1-monocaprate, l-dodecylazacycloheptan-2-one, an acylcarnitine, an acylcholine, or a C 1-10 alkyl ester (e.g., isopropylmyristate IPM), monoglyceride, diglyceride or pharmaceutically acceptable salt thereof. Topical formulations are described in detail in U.S. Patent No. 6,747,014, which is incorporated herein by reference. In addition, dsRNA molecules can be administered to a mammal as biologic or abiologic means as described in, for example, U.S. Pat. No. 6,271,359. Abiologic delivery can be accomplished by a variety of methods including, without limitation, (1) loading liposomes with a dsRNA acid molecule provided herein and (2) complexing a dsRNA molecule with lipids or liposomes to form nucleic acid-lipid or nucleic acid-liposome complexes. The liposome can be composed of cationic and neutral lipids commonly used to transfect cells in vitro. Cationic lipids can complex (e.g., charge-associate) with negatively charged nucleic acids to form liposomes. Examples of cationic liposomes include, without limitation, lipofectin, lipofectamine, lipofectace, and DOTAP. Procedures for forming liposomes are well known in the art. Liposome compositions can be formed, for example, from phosphatidylcholine, dimyristoyl phosphatidylcholine, dipalmitoyl phosphatidylcholine, dimyristoyl phosphatidylglycerol, or dioleoyl phosphatidylethanolamine. Numerous lipophilic agents are commercially available, including Lipofectin™ (Invitrogen/Life Technologies, Carlsbad, Calif.) and Effectene™ (Qiagen, Valencia, Calif.). In addition, systemic delivery methods can be optimized using commercially available cationic lipids such as DDAB or DOTAP, each of which can be mixed with a neutral lipid such as DOPE or cholesterol. In some cases, liposomes such as those described by Templeton et al. (Nature Biotechnology, 15: 647-652 (1997)) can be used. In other embodiments, polycations such as polyethyleneimine can be used to achieve delivery in vivo and ex vivo (Boletta et al. , J. Am Soc. Nephrol. 7: 1728 (1996)). Additional information regarding the use of liposomes to

deliver nucleic acids can be found in U.S. Pat. No. 6,271,359, PCT Publication WO 96/40964 and Morrissey, D. et al. 2005. Nat Biotechnol. 23(8): 1002-7.

Biologic delivery can be accomplished by a variety of methods including, without limitation, the use of viral vectors. For example, viral vectors (e.g., adenovirus and herpesvirus vectors) can be used to deliver dsRNA molecules to liver cells. Standard molecular biology techniques can be used to introduce one or more of the dsRNAs provided herein into one of the many different viral vectors previously developed to deliver nucleic acid to cells. These resulting viral vectors can be used to deliver the one or more dsRNAs to cells by, for example, infection.

Characterization of formulated dsRNAs

Formulations prepared by either the in-line mixing or extrusion-free method can be characterized in similar manners. For example, formulations are typically characterized by visual inspection. They should be whitish translucent solutions free from aggregates or sediment. Particle size and particle size distribution of lipid-nanoparticles can be measured by light scattering using, for example, a Malvern Zetasizer Nano ZS (Malvern, USA). Particles should be about 20-300 nm, such as 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 can be incubated with an RNA-binding dye, such as Ribogreen (Molecular Probes) in the presence or absence of a formulation disrupting surfactant, e.g., 0.5% Triton- XlOO. The total siRNA in the formulation can be 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%. For SNALP formulation, the particle size is at least 30 nm, at least 40 nm, at least 50 nm, at least 60 nm, at least 70 nm, at least 80 nm, at least 90 nm, at least 100 nm, at least 110 nm, and at least 120 nm. The suitable range is typically about at least 50 nm to about at least 110 nm, about at least 60 nm to about at least 100 nm, or about at least 80 nm to about at least 90 nm.

Liposomal formulations

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.

Liposomal 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 ah, 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 ah, 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 is 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 ah, 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 ah, 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™ I (glyceryl dilaurate/cholesterol/po- lyoxyethylene-10-stearyl ether) and Novasome™ 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 MI , 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. Papahadjopoulos et al. (Ann. N.Y. Acad. Sci., 1987, 507, 64) reported the ability of monosialoganglioside G MI , galactocerebroside 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 MI 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, 2Ci 2 i5G, that contains a PEG moiety. Ilium et al (FEBS Lett., 1984, 167, 79) noted that hydrophilic coating 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 Biophysica 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 Bl 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 Bl). 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 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 a dsRNA. 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 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 dissolved 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 sulfates 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 positive 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).

SNALPs

In one embodiment, a dsRNA featured in the invention is fully encapsulated in the lipid formulation to form a SPLP, pSPLP, SNALP, or other nucleic acid-lipid particle. As used herein, the term "SNALP" refers to a stable nucleic acid-lipid particle, including SPLP. As used herein, the term "SPLP" refers to a nucleic acid-lipid particle comprising plasmid

DNA encapsulated within a lipid vesicle. SNALPs and SPLPs typically contain a cationic lipid, a non-cationic lipid, and a lipid that prevents aggregation of the particle (e.g., a PEG- lipid conjugate). SNALPs and SPLPs are extremely useful for systemic applications, as they exhibit extended circulation lifetimes following intravenous (i.v.) injection and accumulate at distal sites (e.g., sites physically separated from the administration site). SPLPs include "pSPLP," which include an encapsulated condensing agent-nucleic acid complex as set forth in PCT Publication No. WO 00/03683. The particles of the present invention typically have a mean diameter of about 50 nm to about 150 nm, more typically about 60 nm to about 130 nm, more typically about 70 nm to about 110 nm, most typically about 70 to about 90 nm, and are substantially nontoxic. In addition, the nucleic acids when present in the nucleic acid- lipid particles of the present invention are resistant in aqueous solution to degradation with a nuclease. Nucleic acid-lipid particles and their method of preparation are disclosed in, e.g., U.S. Patent Nos. 5,976,567; 5,981,501; 6,534,484; 6,586,410; 6,815,432; and PCT Publication No. WO 96/40964.

In one embodiment, the lipid to drug ratio (mass/mass ratio) (e.g., lipid to dsRNA ratio) will be in the range of from about 1 : 1 to about 50: 1, from about 1 : 1 to about 25: 1, from about 3: 1 to about 15: 1, from about 4: 1 to about 10: 1, from about 5: 1 to about 9: 1, or about 6: 1 to about 9: 1.

The cationic lipid may be, for example, N,N-dioleyl-N,N-dimethylammonium chloride (DODAC), N,N-distearyl-N,N-dimethylammonium bromide (DDAB), N-(I -(2,3- dioleoyloxy)propyl)-N,N,N-trimethylammonium chloride (DOTAP), N-(I -(2,3- dioleyloxy)propyl)-N,N,N-trimethylammonium chloride (DOTMA), N,N-dimethyl-2,3- dioleyloxy propylamine (DODMA), 1 ,2-DiLinoleyloxy-N,N-dimethylaminopropane (DLinDMA), l,2-Dilinolenyloxy-N,N-dimethylaminopropane (DLenDMA), 1,2- Dilinoleylcarbamoyloxy-3-dimethylaminopropane (DLin-C-DAP), 1 ,2-Dilinoleyoxy-3- (dimethylamino)acetoxypropane (DLin-DAC), 1 ,2-Dilinoleyoxy-3 -morpholinopropane (DLin-MA), l,2-Dilinoleoyl-3-dimethylaminopropane (DLinDAP), l,2-Dilinoleylthio-3- dimethylaminopropane (DLin-S-DMA), 1 -Linoleoyl-2-linoleyloxy-3 -dimethylaminopropane (DLin-2-DMAP), l,2-Dilinoleyloxy-3-trimethylaminopropane chloride salt (DLin-TMA.Cl), l,2-Dilinoleoyl-3-trimethylaminopropane chloride salt (DLin-TAP.Cl), l,2-Dilinoleyloxy-3- (N-methylpiperazino)propane (DLin-MPZ), or 3-(N,N-Dilinoleylamino)-l,2-propanediol (DLinAP), 3-(N,N-Dioleylamino)-l,2-propanedio (DOAP), l,2-Dilinoleyloxo-3-(2-N,N- dimethylamino)ethoxypropane (DLin-EG-DMA), 2,2-Dilinoleyl-4-dimethylaminomethyl-

[l,3]-dioxolane (DLin-K-DMA) or analogs thereof, or a mixture thereof. The cationic lipid may comprise from about 20 mol % to about 50 mol % or about 40 mol % of the total lipid present in the particle.

In another embodiment, the compound 2,2-Dilinoleyl-4-dimethylaminoethyl-[l,3]- dioxolane can be used to prepare lipid-siRNA nanoparticles. Synthesis of 2,2-Dilinoleyl-4- dimethylaminoethyl-[l,3]-dioxolane is described in United States provisional patent application number 61/107,998 filed on October 23, 2008, which is herein incorporated by reference.

In one embodiment, the lipid-siRNA particle includes 40% 2-Dilinoleyl-4- dimethylaminoethyl-[l,3]-dioxolane: 10% DSPC: 40% Cholesterol: 10% PEG-C-DOMG (mole percent) with a particle size of 63.0 ± 20 nm and a 0.027 siRNA/Lipid Ratio.

The non-cationic lipid may be an anionic lipid or a neutral lipid including, but not limited to, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl-phosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl- phosphatidylethanolamine (POPE), dioleoyl- phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane-l- carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1 -trans PE, 1 -stearoyl-2-oleoyl- phosphatidyethanolamine (SOPE), cholesterol, or a mixture thereof. The non-cationic lipid may be from about 5 mol % to about 90 mol %, about 10 mol %, or about 58 mol % if cholesterol is included, of the total lipid present in the particle.

The conjugated lipid that inhibits aggregation of particles may be, for example, a polyethyleneglycol (PEG)-lipid including, without limitation, a PEG-diacylglycerol (DAG), a PEG-dialkyloxypropyl (DAA), a PEG-phospholipid, a PEG-ceramide (Cer), or a mixture thereof. The PEG-DAA conjugate may be, for example, a PEG-dilauryloxypropyl (Ci 2 ), a PEG-dimyristyloxypropyl (Ci 4 ), a PEG-dipalmityloxypropyl (Ciβ), or a PEG- distearyloxypropyl (C] 8 ). The conjugated lipid that prevents aggregation of particles may be from 0 mol % to about 20 mol % or about 2 mol % of the total lipid present in the particle.

In some embodiments, the nucleic acid-lipid particle further includes cholesterol at, e.g., about 10 mol % to about 60 mol % or about 48 mol % of the total lipid present in the particle.

LNPOl

In one embodiment, the lipidoid ND98-4HC1 (MW 1487) (Formula 1), Cholesterol (Sigma- Aldrich), and PEG-Ceramide C 16 (Avanti Polar Lipids) can be used to prepare lipid- siRNA nanoparticles (i.e., LNPOl particles). Stock solutions of each in ethanol can be prepared as follows: ND98, 133 mg/ml; Cholesterol, 25 mg/ml, PEG-Ceramide C16, 100 mg/ml. The ND98, Cholesterol, and PEG-Ceramide C 16 stock solutions can then be combined in a, e.g., 42:48: 10 molar ratio. The combined lipid solution can be mixed with aqueous siRNA (e.g., in sodium acetate pH 5) such that the final ethanol concentration is about 35-45% and the final sodium acetate concentration is about 100-300 mM. Lipid- siRNA nanoparticles typically form spontaneously upon mixing. Depending on the desired particle size distribution, the resultant nanoparticle mixture can be extruded through a polycarbonate membrane (e.g., 100 nm cut-off) using, for example, a thermobarrel extruder, such as Lipex Extruder (Northern Lipids, Inc). In some cases, the extrusion step can be omitted. Ethanol removal and simultaneous buffer exchange can be accomplished by, for example, dialysis or tangential flow filtration. Buffer can be exchanged with, for example, phosphate buffered saline (PBS) at about pH 7, e.g., about pH 6.9, about pH 7.0, about pH 7.1, about pH 7.2, about pH 7.3, or about pH 7.4.

Formula 1

LNPOl formulations are described, e.g., in International Application Publication No. WO 2008/042973, which is hereby incorporated by reference.

Emulsions

The compositions of the present invention may be prepared and formulated as emulsions. Emulsions are typically heterogeneous systems of one liquid dispersed in another in the form of droplets usually exceeding 0.1 μm in diameter (Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199; 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 2, p. 335; Higuchi et ah, 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, Marcel 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, volume

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 emulsifϊers 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 semisolid 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 emulsion 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 Banker (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 external 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, quaternary ammonium salts,

benzalkonium chloride, esters of p-hydroxybenzoic acid, and boric acid. Antioxidants are also commonly added to emulsion formulations to prevent deterioration 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 metabisulfite, 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 molecules (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, polyoxyethylene oleyl ethers, polyglycerol 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 cosurfactant, 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 acid 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. Lipid based microemulsions (both o/w and w/o) have been proposed to enhance the oral bioavailability of drugs, including peptides (Constantinides 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 solid dosage forms, improved clinical potency, and decreased toxicity (Constantinides et ah, 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, peptides 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 ah, Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92). Each of these classes has been discussed above.

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 perfluorochemical 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, l-dodecylazacycloheptan-2-one, acylcarnitines, acylcholines, C 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 ah, Critical Reviews in Therapeutic Drug Carryier Systems, 1991, p.92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33; El Hariri et ah, 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. Suitable bile salts 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), glycodeoxycholic acid (sodium glycodeoxycholate), taurocholic acid (sodium taurocholate), taurodeoxycholic acid (sodium taurodeoxycholate), chenodeoxycholic acid (sodium chenodeoxycholate), ursodeoxycholic acid (UDCA), sodium tauro-24,25-dihydro-fusidate (STDHF), sodium glycodihydrofusidate and polyoxyethylene-9-lauryl ether (POE) (Lee et ah, 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, 7, 1- 33; Yamamoto et ah, J. Pharm. Exp. Ther., 1992, 263, 25; Yamashita et ah, J. Pharm. Sci., 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). Suitable chelating agents include but are not limited to disodium ethylenediaminetetraacetate (EDTA), citric acid, salicylates (e.g., sodium salicylate, 5 -methoxy salicylate and homovanilate), N-acyl derivatives of collagen, laureth-9 and N-amino acyl derivatives of beta-diketones (enamines)(Lee et ah, Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33; Buur et ah, 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 mucosa (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 -alkenylazacyclo-alkanone derivatives (Lee et ah, 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 ah, 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 lipofectin (Junichi et al, U.S. Pat. No. 5,705,188), cationic glycerol derivatives, and polycationic molecules, such as poly lysine (Lollo et ah, 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 dsRNAs of the present invention can be formulated in a pharmaceutically acceptable carrier or diluent. A "pharmaceutically acceptable carrier" (also referred to herein as an "excipient") is a pharmaceutically acceptable solvent, suspending agent, or any other

pharmacologically inert vehicle. Pharmaceutically acceptable carriers can be liquid or solid, and can be selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, and other pertinent transport and chemical properties. Typical pharmaceutically acceptable carriers include, by way of example and not limitation: water; saline solution; binding agents (e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose and other sugars, gelatin, or calcium sulfate); lubricants (e.g., starch, polyethylene glycol, or sodium acetate); disintegrates (e.g., starch or sodium starch glycolate); and wetting agents (e.g., sodium lauryl sulfate).

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 co-administration 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 extra- circulatory 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 co-administered with polyinosinic acid, dextran sulfate, polycytidic acid or 4-acetamido-4'isothiocyano-stilbene- 2,2'-disulfonic acid (Miyao et ah, DsRNA Res. Dev., 1995, 5, 115-121; Takakura et ah, 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 solid 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, talc, 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 excipients 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 topical 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, antipruritics, 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.

Combination therapy

In one aspect, a composition of the invention can be used in combination therapy. The term "combination therapy" includes the administration of the subject compounds in further combination with other biologically active ingredients (such as, but not limited to, a second and different antineoplastic agent) and non-drug therapies (such as, but not limited to, surgery or radiation treatment). For instance, the compounds of the invention can be used in combination with other pharmaceutically active compounds, preferably compounds that are able to enhance the effect of the compounds of the invention. The compounds of the invention can be administered simultaneously (as a single preparation or separate preparation) or sequentially to the other drug therapy. In general, a combination therapy envisions administration of two or more drugs during a single cycle or course of therapy.

In one aspect of the invention, the subject compounds may be administered in combination with one or more separate agents that modulate protein kinases involved in various disease states. Examples of such kinases may include, but are not limited to: serine/threonine specific kinases, receptor tyrosine specific kinases and non-receptor tyrosine specific kinases. Serine/threonine kinases include mitogen activated protein kinases (MAPK), meiosis specific kinase (MEK), RAF and aurora kinase. Examples of receptor kinase families include epidermal growth factor receptor (EGFR) (e.g., HER2/neu, HER3, HER4, ErbB, ErbB2, ErbB3, ErbB4, Xmrk, DER, Let23); fibroblast growth factor (FGF) receptor (e.g. FGF-Rl, GFF-R2/BEK/CEK3, FGF-R3/CEK2, FGF-R4/TKF, KGF-R); hepatocyte growth/scatter factor receptor (HGFR) (e.g., MET, RON, SEA, SEX); insulin receptor (e.g. IGFI-R); Eph (e.g. CEK5, CEK8, EBK, ECK, EEK, EHK-I, EHK-2, ELK, EPH, ERK, HEK, MDK2, MDK5, SEK); AxI (e.g. Mer/Nyk, Rse); RET; and platelet- derived growth factor receptor (PDGFR) (e.g. PDGFα-R, PDGβ-R, CSFl -R/FMS, SCF- R/C-KIT, VEGF-R/FLT, NEK/FLKl, FLT3/FLK2/STK-1). Non-receptor tyrosine kinase families include, but are not limited to, BCR-ABL (e.g. p43 abl , ARG); BTK (e.g. ITK/EMT, TEC); CSK, FAK, FPS, JAK, SRC, BMX, FER, CDK and SYK.

In another aspect of the invention, the subject compounds may be administered in combination with one or more agents that modulate non-kinase biological targets or processes. Such targets include histone deacetylases (HDAC), DNA methyltransferase (DNMT), heat shock proteins (e.g., HSP90), and proteosomes.

In one embodiment, subject compounds may be combined with antineoplastic agents (e.g. small molecules, monoclonal antibodies, antisense RNA, and fusion proteins) that inhibit one or more biological targets such as Zolinza, Tarceva, Iressa, Tykerb, Gleevec,

Sutent, Sprycel, Nexavar, Sorafenib, CNF2024, RG108, BMS387032, Affmitak, Avastin, Herceptin, Erbitux, AG24322, PD325901 , ZD6474, PD 184322, Obatodax, ABT737 and AEE788. Such combinations may enhance therapeutic efficacy over efficacy achieved by any of the agents alone and may prevent or delay the appearance of resistant mutational variants.

In certain preferred embodiments, the compounds of the invention are administered in combination with a chemotherapeutic agent. Chemotherapeutic agents encompass a wide range of therapeutic treatments in the field of oncology. These agents are administered at various stages of the disease for the purposes of shrinking tumors, destroying remaining cancer cells left over after surgery, inducing remission, maintaining remission and/or alleviating symptoms relating to the cancer or its treatment. Examples of such agents include, but are not limited to, alkylating agents such as mustard gas derivatives (Mechlorethamine, cylophosphamide, chlorambucil, melphalan, ifosfamide), ethylenimines (thiotepa, hexamethylmelanine), Alkylsulfonates (Busulfan), Hydrazines and Triazines (Altretamine, Procarbazine, Dacarbazine and Temozolomide), Nitrosoureas (Carmustine, Lomustine and Streptozocin), Ifosfamide and metal salts (Carboplatin, Cisplatin, and Oxaliplatin); plant alkaloids such as Podophyllotoxins (Etoposide and Tenisopide), Taxanes (Paclitaxel and Docetaxel), Vinca alkaloids (Vincristine, Vinblastine, Vindesine and Vinorelbine), and Camptothecan analogs (Irinotecan and Topotecan); anti-tumor antibiotics such as Chromomycins (Dactinomycin and Plicamycin), Anthracyclines (Doxorubicin, Daunorubicin, Epirubicin, Mitoxantrone, Valrubicin and Idarubicin), and miscellaneous antibiotics such as Mitomycin, Actinomycin and Bleomycin; anti-metabolites such as folic acid antagonists (Methotrexate, Pemetrexed, Raltitrexed, Aminopterin), pyrimidine antagonists (5-Fluorouracil, Floxuridine, Cytarabine, Capecitabine, and Gemcitabine), purine antagonists (6-Mercaptopurine and 6-Thioguanine) and adenosine deaminase inhibitors (Cladribine, Fludarabine, Mercaptopurine, Clofarabine, Thioguanine, Nelarabine and Pentostatin); topoisomerase inhibitors such as topoisomerase I inhibitors (Ironotecan, topotecan) and topoisomerase II inhibitors (Amsacrine, etoposide, etoposide phosphate, teniposide); monoclonal antibodies (Alemtuzumab, Gemtuzumab ozogamicin, Rituximab, Trastuzumab, Ibritumomab Tioxetan, Cetuximab, Panitumumab, Tositumomab, Bevacizumab); and miscellaneous anti-neoplasties such as ribonucleotide reductase inhibitors (Hydroxyurea); adrenocortical steroid inhibitor (Mitotane); enzymes (Asparaginase and Pegaspargase); anti-microtubule agents (Estramustine); and retinoids (Bexarotene, Isotretinoin, Tretinoin (ATRA). In certain preferred embodiments, the compounds of the invention are administered in combination with a chemoprotective agent. Chemoprotective

agents act to protect the body or minimize the side effects of chemotherapy. Examples of such agents include, but are not limited to, amfostine, mesna, and dexrazoxane.

In one aspect of the invention, the subject compounds are administered in combination with radiation therapy. Radiation is commonly delivered internally (implantation of radioactive material near cancer site) or externally from a machine that employs photon (x- ray or gamma-ray) or particle radiation. Where the combination therapy further comprises radiation treatment, the radiation treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and radiation treatment is achieved. For example, in appropriate cases, the beneficial effect is still achieved when the radiation treatment is temporally removed from the administration of the therapeutic agents, perhaps by days or even weeks.

It will be appreciated that compounds of the invention can be used in combination with an immunotherapeutic agent. One form of immunotherapy is the generation of an active systemic tumor-specific immune response of host origin by administering a vaccine composition at a site distant from the tumor. Various types of vaccines have been proposed, including isolated tumor-antigen vaccines and anti-idiotype vaccines. Another approach is to use tumor cells from the subject to be treated, or a derivative of such cells (reviewed by Schirrmacher et al. (1995) J. Cancer Res. Clin. Oncol. 121 :487). In U.S. Pat. No. 5,484,596, Hanna Jr. et al. claim a method for treating a resectable carcinoma to prevent recurrence or metastases, comprising surgically removing the tumor, dispersing the cells with collagenase, irradiating the cells, and vaccinating the patient with at least three consecutive doses of about 10 7 cells.

It will be appreciated that the compounds of the invention may advantageously be used in conjunction with one or more adjunctive therapeutic agents. Examples of suitable agents for adjunctive therapy include steroids, such as corticosteroids (amcinonide, betamethasone, betamethasone dipropionate, betamethasone valerate, budesonide, clobetasol, clobetasol acetate, clobetasol butyrate, clobetasol 17-propionate, cortisone, deflazacort, desoximetasone, diflucortolone valerate, dexamethasone, dexamethasone sodium phosphate, desonide, furoate, fluocinonide, fluocinolone acetonide, halcinonide, hydrocortisone, hydrocortisone butyrate, hydrocortisone sodium succinate, hydrocortisone valerate, methyl prednisolone, mometasone, prednicarbate, prednisolone, triamcinolone, triamcinolone acetonide, and halobetasol proprionate); a 5HTi agonist, such as a triptan (e.g. sumatriptan or naratriptan); an adenosine Al agonist; an EP ligand; an NMDA modulator, such as a glycine antagonist; a sodium channel blocker (e.g. lamotrigine); a substance P antagonist (e.g. an NKi

antagonist); a cannabinoid; acetaminophen or phenacetin; a 5 -lipoxygenase inhibitor; a leukotriene receptor antagonist; a DMARD (e.g. methotrexate); gabapentin and related compounds; a tricyclic antidepressant (e.g. amitryptilline); a neurone stabilizing antiepileptic drug; a mono-aminergic uptake inhibitor (e.g. venlafaxine); a matrix metalloproteinase inhibitor; a nitric oxide synthase (NOS) inhibitor, such as an iNOS or an nNOS inhibitor; an inhibitor of the release, or action, of tumour necrosis factor α; an antibody therapy, such as a monoclonal antibody therapy; an antiviral agent, such as a nucleoside inhibitor (e.g. lamivudine) or an immune system modulator (e.g. interferon); an opioid analgesic; a local anaesthetic; a stimulant, including caffeine; an H2-antagonist (e.g. ranitidine); a proton pump inhibitor (e.g. omeprazole); an antacid (e.g. aluminium or magnesium hydroxide; an antiflatulent (e.g. simethicone); a decongestant (e.g. phenylephrine, phenylpropanolamine, pseudoephedrine, oxymetazoline, epinephrine, naphazoline, xylometazoline, propylhexedrine, or levo-desoxyephedrine); an antitussive (e.g. codeine, hydrocodone, carmiphen, carbetapentane, or dextramethorphan); a diuretic; or a sedating or non-sedating antihistamine.

The compounds of the invention can be co-administered with siRNA that target other genes. For example, a compound of the invention can be co-administered with an siRNA targeted to a c-Myc gene. In one example, AD- 12115 can be co-administered with a c-Myc siRNA. Examples of c-Myc targeted siRNAs are disclosed in United States patent application number 12/373,039 which is herein incorporated by reference.

Methods for treating diseases caused by expression of the Eg5 and VEGF genes The invention relates in particular to the use of a composition containing at least two dsRNAs, one targeting an Eg5 gene, and one targeting a VEGF gene, for the treatment of a cancer, such as liver cancer, e.g., for inhibiting tumor growth and tumor metastasis. For example, a composition, such as pharmaceutical composition, may be used for the treatment of solid tumors, like intrahepatic tumors such as may occur in cancers of the liver. A composition containing a dsRNA targeting Eg5 and a dsRNA targeting VEGF may also be used to treat other tumors and cancers, such as breast cancer, lung cancer, head and neck cancer, brain cancer, abdominal cancer, colon cancer, colorectal cancer, esophagus cancer, gastrointestinal cancer, glioma, 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 lymphomas and blood cancer. The invention further relates to the use of a composition containing an Eg5 dsRNA and a VEGF dsRNA for inhibiting accumulation of ascites fluid and pleural effusion in

different types of cancer, e.g., liver cancer, breast cancer, lung cancer, head cancer, neck cancer, brain cancer, abdominal cancer, colon cancer, colorectal cancer, esophagus cancer, gastrointestinal cancer, glioma, 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 effects on Eg5 and VEGF expression, a composition according to the invention or a pharmaceutical composition prepared therefrom can enhance the quality of life.

In one embodiment, a patient having a tumor associated with AFP expression, or a tumor secreting AFP, e.g., a hepatoma or teratoma, is treated. In certain embodiments, the patient has a malignant teratoma, an endodermal sinus tumor (yolk sac carcinoma), a neuroblastoma, a hepatoblastoma, a heptocellular carcinoma, testicular cancer or ovarian cancer.

The invention furthermore relates to the use of a 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-fluorouracil, adriamycin, daunorubicin or tamoxifen.

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. 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, taxol 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 and the VEGF gene

In yet another aspect, the invention provides a method for inhibiting the expression of the Eg5 gene and the VEGF gene in a mammal. The method includes administering a composition featured in the invention to the mammal such that expression of the target Eg5 gene and the target VEGF gene is silenced.

In one embodiment, a method for inhibiting Eg5 gene expression and VEGF gene expression includes administering a composition containing two different dsRNA molecules, one having a nucleotide sequence that is complementary to at least a part of an RNA transcript of the Eg5 gene and the other having a nucleotide sequence that is complementary to at least a part of an RNA transcript of the VEGF gene of the mammal to be treated. When the organism to be treated is a mammal such as a human, the composition 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

Example 1. 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

For screening of dsRNA, 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, 5OθA, 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 Biochemie 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 described in Current protocols in nucleic acid chemistry, Beaucage, S.L. et al. (Edrs.), John Wiley & Sons, Inc., New York, NY, 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, Unterschleifiheim, 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), heated in a water bath at 85 - 90 0 C for 3 minutes and cooled to room temperature over a period of 3 - 4 hours. The annealed RNA solution was stored at -20 0 C until use.

Conjugates

The following is a prophetic description of 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

A 4.7 M aqueous solution of sodium hydroxide (50 mL) was added into a stirred, ice- cooled solution of ethyl 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 x 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-a mino)-hexanoyl]- amino} -propionic acid ethyl ester AB

AB

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 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 urea. 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%) ofAB.

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

AC

3-{Ethoxycarbonylmethyl-[6-(9H-fluoren-9-ylmethoxycarbonylam ino)-hexanoyl]- amino} -propionic acid ethyl ester AB (11.5 g, 21.3 mmol) was dissolved in 20% piperidine in dimethylformamide at 0 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-hexylH0,13-dimethyl-2,3,4,7,8,9,10,l l, 12,13,14,15,16,17- tetradecahydro-lH-cyclopenta[a]phenanthren-3-yloxycarbonylam ino]- hexanoyl}ethoxycarbonylmethyl-amino)-propionic acid ethyl ester AD

AD

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

AE

Potassium t-butoxide (1.1 g, 9.8 mmol) was slurried in 30 mL of dry toluene. The mixture was cooled to 0 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 0 C and 1 mL of glacial acetic acid was added, immediately followed by 4 g of NaH 2 PO 4 -H 2 O 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 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-ketoester (39%).

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

AF

Methanol (2 mL) 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 HCl (12.5 mL) was added, the mixture was extracted with ethylacetate (3 x 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/CHCl 3 ) (89%).

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

AG

Diol AF (1.25 gm 1.994 mmol) was dried by evaporating with pyridine (2 x 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 IM 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/CHCl 3 ) (1.75 g, 95%).

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

AH

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 0 C overnight. The mixture was dissolved in anhydrous dichloroethane (3 mL), triethylamine (0.318 g, 0.440 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 X 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

AI

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/pyridine. 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. dsRNA targeting the Eg5 gene

Initial Screening set siRNA design was carried out to identify siRNAs targeting Eg5 (also known as KIFl 1, HSKP, KNSLl and TRIP5). Human mRNA sequences to Eg5, RefSeq ID numbeπNM 004523, was used.

siRNA duplexes cross-reactive to human and mouse Eg5 were designed. Twenty-four duplexes were synthesized for screening. (Table Ia). A second screening set was defined with 266 siRNAs targeting human Eg5, as well as its rhesus monkey ortholog (Table 2a). 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 a).

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:NM_004523.2, 4908 bp, and Rhesus EG5: XM_001087644.1, 878 bp (only 5' part of human EG5)

For the Tables: Key: A,G,C,U-ribonucleotides: T-deoxythymidine: u,c-2'-O-methyl nucleotides: s-phosphorothioate linkage.

Table Ia. Sequences of Eg5/ KSP dsRNA duplexes

Table Ib. Analysis of Eg5/KSP ds duplexes single dose screen @

25 nM [ % SDs 2nd screen duplex residual (among name mRNA] quadruplicates )

AL-DP-6226 23% 3%

AL-DP-6227 69% 10%

AL-DP-6228 33% 2%

AL-DP-6229 2% 2%

AL-DP-6230 66% 11 %

AL-DP-6231 17% 1 %

AL-DP-6232 9% 3%

AL-DP-6233 24% 6%

AL-DP-6234 91 % 2%

AL-DP-6235 1 12% 4%

AL-DP-6236 69% 4%

AL-DP-6237 42% 2%

AL-DP-6238 45% 2%

AL-DP-6239 2% 1 %

AL-DP-6240 48% 2%

AL-DP-6241 41 % 2%

AL-DP-6242 8% 2%

AL-DP-6243 7% 1 %

AL-DP-6244 6% 2%

AL-DP-6245 12% 2%

AL-DP-6246 28% 3%

AL-DP-6247 71 % 4%

AL-DP-6248 5% 2%

AL-DP-6249 28% 3%

Table 2a. Sequences of Eg5/ KSP dsRNA duplexes

EQ SEQ SEQ sequence of 19-mer antisense sequence (5 - duplex

[D ID sense sequence (5 -3 ) target site ) ID

3 ) name

0 : NO. NO.

'68 CAUACUCUAGUCGUUCCCA 49 cAuAcucuAGucGuucccAT: T 50 UGGGAACGACuAGAGuAUGTsT AD-12072

'69 AGCGCCCAUUCAAUAGUAG 51 AGcGcccAuucAAuAGuAGT: T 52 CuACuAUUGAAUGGGCGCUTsT AD-12073

'70 GGAAAGCUAGCGCCCAUUC 53 GGAAAGcuAGcGcccAuucT: T 54 GAAUGGGCGCuAGCUUUCCTsT AD-1207 4

'71 GAAAGCUAGCGCCCAUUCA 55 GAAAGcuAGcGcccAuucAT: T 56 UGAAUGGGCGCuAGCUUUCTsT AD-12075

'72 AGAAACUACGAUUGAUGGA 57 AGAAAcuAcGAuuGAuGGAT: T 58 UCcAUcAAUCGuAGUUUCUTsT AD-12076

'73 UGUUCCUUAUCGAGAAUCU 59 uGuuccuuAucGAGAAucuT: T 60 AGAUUCUCGAuAAGGAAcATsT AD-12077

'74 CAGAUUACCUCUGCGAGCC 61 cAGAuuAccucuGcGAGccT: T 62 GGCUCGcAGAGGuAAUCUGTsT AD-12078

'75 GCGCCCAUUCAAUAGUAGA 63 GcGcccAuucAAuAGuAGAT: T 64 UCuACuAUUGAAUGGGCGCTsT AD-12079

'76 UUGCACUAUCUUUGCGUAU 65 uuGcAcuAucuuuGcGuAuT: T 66 AuACGcAAAGAuAGUGcAATsT AD-12080

'77 CAGAGCGGAAAGCUAGCGC 67 cAGAGcGGAAAGcuAGcGcT: T 68 GCGCuAGCUUUCCGCUCUGTsT AD-12081

'78 AGACCUUAUUUGGUAAUCU 69 AGAccuuAuuuGGuAAucuT: T 70 AGAUuACcAAAuAAGGUCUTsT AD-12082

'79 AUUCUCUUGGAGGGCGUAC 71 AuucucuuGGAGGGcGuAcT: T 72 GuACGCCCUCcAAGAGAAUTsT AD-12083

'80 GGCUGGUAUAAUUCCACGU 73 GGcuGGuAuAAuuccAcGuT: T 74 ACGUGGAAUuAuACcAGCCTsT AD-1208 4

'81 GCGGAAAGCUAGCGCCCAU 75 GcGGAAAGcuAGcGcccAuT: T 76 AUGGGCGCuAGCUUUCCGCTsT AD-12085

'82 UGCACUAUCUUUGCGUAUG 77 uGcAcuAucuuuGcGuAuGT: T 78 cAuACGcAAAGAuAGUGcATsT AD-12086

'83 GUAUAAUUCCACGUACCCU 79 GuAuAAuuccAcGuAcccuT: T 80 AGGGuACGUGGAAUuAuACTsT AD-12087

'84 AGAAUCUAAACUAACUAGA 81 AGAAuCuAAAcuAAcuAGAT: T 82 UCuAGUuAGUUuAGAUUCUTsT AD-12088

'85 AGGAGCUGAAUAGGGUUAC 83 AGGAGcuGAAuAGGGuuAcT: T 84 GuAACCCuAUUcAGCUCCUTsT AD-12089

'86 GAAGUACAUAAGACCUUAU 85 GAAGuAcAuAAGAccuuAuT: T 86 AuAAGGUCUuAUGuACUUCTsT AD-12090

'87 GACAGUGGCCGAUAAGAUA 87 GAcAGuGGccGAuAAGAuAT: T 88 uAUCUuAUCGGCcACUGUCTsT AD-12091

'88 AAACCACUUAGUAGUGUCC 89 AAAccAcuuAGuAGuGuccT: T 90 GGAcACuACuAAGUGGUUUTsT AD-12092

'89 UCCCUAGACUUCCCUAUUU 91 ucccuAGAcuucccuAuuuT: T 92 AAAuAGGGAAGUCuAGGGATsT AD-12093

'90 UAGACUUCCCUAUUUCGCU 93 uAGAcuucccuAuuucGcuT: T 94 AGCGAAAuAGGGAAGUCuATsT AD-1209 4

'91 GCGUCGCAGCCAAAUUCGU 95 GcGucGcAGccAAAuucGuT: T 96 ACGAAUUUGGCUGCGACGCTST AD-12095

'92 AGCUAGCGCCCAUUCAAUA 97 AGcuAGcGcccAuucAAuAT: T 98 uAUUGAAUGGGCGCuAGCUTsT AD-12096

'93 GAAACUACGAUUGAUGGAG 99 GAAAcuAcGAuuGAuGGAGT: T 100 CUCcAUcAAUCGuAGUUUCTsT AD-12097

'94 CCGAUAAGAUAGAAGAUCA 101 CCGAuAAGAuAGAAGAuCAT: T 102 UGAUCUUCuAUCUuAUCGGTsT AD-12098

'95 UAGCGCCCAUUCAAUAGUA 103 uAGcGcccAuucAAuAGuAT: T 104 uACuAUUGAAUGGGCGCuATsT AD-12099

'96 UUUGCGUAUGGCCAAACUG 105 uuuGcGuAuGGccAAAcuGT: T 106 cAGUUUGGCcAuACGcAAATsT AD-12100

'97 CACGUACCCUUCAUCAAAU 107 cAcGuAcccuucAucAAAuT: T 108 AUUUGAUGAAGGGUACGUGTST AD-12101

'98 UCUUUGCGUAUGGCCAAAC 109 ucuuuGcGuAuGGccAAAcT: T 110 GUUUGGCcAuACGcAAAGATsT AD-12102

'99 CCGAAGUGUUGUUUGUCCA 111 ccGAAGuGuuGuuuGuccAT: T 112 UGGAcAAAcAAcACUUCGGTsT AD-12103

300 AGAGCGGAAAGCUAGCGCC 113 AGAGcGGAAAGcuAGcGccT: T 114 GGCGCuAGCUUUCCGCUCUTsT AD-1210 4

301 GCUAGCGCCCAUUCAAUAG 115 GcuAGcGcccAuucAAuAGT: T 116 CuAUUGAAUGGGCGCuAGCTsT AD-12105 AAGUUAGUGUACGAACUGG 117 AAGuuAGuGuAcGAAcuGGT: T 118 CcAGUUCGuAcACuAACUUTsT AD-12106

303 GUACGAACUGGAGGAUUGG 119 GuAcGAAcuGGAGGAuuGGT: T 120 CcAAUCCUCcAGUUCGuACTsT AD-12107 304 ACGAACUGGAGGAUUGGCU 121 AcGAAcuGGAGGAuuGGcuT: T 122 AGCcAAUCCUCcAGUUCGUTsT AD-12108 305 AGAUUGAUGUUUACCGAAG 123 AGAuuGAuGuuuAccGAAGT: T 124 CUUCGGuAAAcAUcAAUCUTsT AD-12109 306 UAUGGGCUAUAAUUGCACU 125 uAuGGGcuAuAAuuGcAcuT: T 126 AGUGcAAUuAuAGCCcAuATsT AD-12110 307 AUCUUUGCGUAUGGCCAAA 127 AucuuuGcGuAuGGccAAAT: T 128 UUUGGCcAuACGcAAAGAUTsT AD-12111 308 ACUCUAGUCGUUCCCACUC 129 AcucuAGucGuucccAcucT: T 130 GAGUGGGAACGACUAGAGUTST AD-12112 309 AACUACGAUUGAUGGAGAA 131 AAcuAcGAuuGAuGGAGAAT: T 132 UUCUCcAUcAAUCGuAGUUTsT AD-12113 310 GAUAAGAGAGCUCGGGAAG 133 GAuAAGAGAGcucGGGAAGT: T 134 CUUCCCGAGCUCUCUUAUCTST AD-1211 4 311 UCGAGAAUCUAAACUAACU 135 ucGAGAAuCuAAAcuAAcuT: T 136 AGUuAGUUuAGAUUCUCGATsT AD-12115 312 AACUAACUAGAAUCCUCCA 137 AAcuAAcuAGAAuCCUCCAT: T 138 UGGAGGAUUCuAGUuAGUUTsT AD-12116 313 GGAUCGUAAGAAGGCAGUU 139 GGAucGuAAGAAGGcAGuuT: T 140 AACUGCCUUCUuACGAUCCTsT AD-12117 314 AUCGUAAGAAGGCAGUUGA 141 AucGuAAGAAGGcAGuuGAT: T 142 UcAACUGCCUUCUuACGAUTsT AD-12118 315 AGGCAGUUGACCAACACAA 143 AGGcAGuuGAccAAcAcAAT: T 144 UUGUGUUGGUcAACUGCCUTsT AD-12119 316 UGGCCGAUAAGAUAGAAGA 145 uGGccGAuAAGAuAGAAGAT: T 146 UCUUCuAUCUuAUCGGCcATsT AD-12120 317 UCUAAGGAUAUAGUCAACA 147 uCuAAGGAuAuAGucAAcAT: T 148 UGUUGACuAuAUCCUuAGATsT AD-12121 318 ACUAAGCUUAAUUGCUUUC 149 AcuAAGcuuAAuuGcuuucT: T 150 GAAAGcAAUuAAGCUuAGUTsT AD-12122 319 GCCCAGAUCAACCUUUAAU 151 GcccAGAucAAccuuuAAuT: T 152 AUuAAAGGUUGAUCUGGGCTsT AD-12123 320 UUAAUUUGGCAGAGCGGAA 153 uuAAuuuGGcAGAGcGGAAT: T 154 UUCCGCUCUGCcAAAUuAATsT AD-1212 4 321 UUAUCGAGAAUCUAAACUA 155 uuAucGAGAAuCuAAAcuAT: T 156 uAGUUuAGAUUCUCGAuAATsT AD-12125 322 CUAGCGCCCAUUCAAUAGU 157 cuAGcGcccAuucAAuAGuT: T 158 ACuAUUGAAUGGGCGCuAGTsT AD-12126 323 AAUAGUAGAAUGUGAUCCU 159 AAuAGuAGAAuGuGAuCCuT: T 160 AGGAUcAcAUUCuACuAUUTsT AD-12127 324 UACGAAAAGAAGUUAGUGU 161 uAcGAAAAGAAGuuAGuGuT: T 162 AcACuAACUUCUUUUCGuATsT AD-12128 325 AGAAGUUAGUGUACGAACU 163 AGAAGuuAGuGuAcGAAcuT: T 164 AGUUCGuAcACuAACUUCUTsT AD-12129 326 ACUAAACAGAUUGAUGUUU 165 AcuAAAcAGAuuGAuGuuuT: T 166 AAAcAUcAAUCUGUUuAGUTsT AD-12130 327 CUUUGCGUAUGGCCAAACU 167 cuuuGcGuAuGGccAAAcuT: T 168 AGUUUGGCcAuACGcAAAGTsT AD-12131 328 AAUGAAGAGUAUACCUGGG 169 AAuGAAGAGuAuAccuGGGT: T 170 CCcAGGuAuACUCUUcAUUTsT AD-12132 329 AUAAUUCCACGUACCCUUC 171 AuAAuuccAcGuAcccuucT: T 172 GAAGGGuACGUGGAAUuAUTsT AD-12133 330 ACGUACCCUUCAUCAAAUU 173 AcGuAcccuucAucAAAuuT: T 174 AAUUUGAUGAAGGGUACGUTST AD-1213 4 331 CGUACCCUUCAUCAAAUUU 175 cGuAcccuucAucAAAuuuT: T 176 AAAUUUGAUGAAGGGUACGTST AD-12135 332 GUACCCUUCAUCAAAUUUU 177 GuAcccuucAucAAAuuuuT: T 178 AAAAUUUGAUGAAGGGUACTST AD-12136 333 AACUUACUGAUAAUGGUAC 179 AAcuuAcuGAuAAuGGuAcT: T 180 GuACcAUuAUcAGuAAGUUTsT AD-12137 334 UUCAGUCAAAGUGUCUCUG 181 uucAGucAAAGuGucucuGT: T 182 cAGAGAcACUUUGACUGAATsT AD-12138 335 UUCUUAAUCCAUCAUCUGA 183 uucuuAAuccAucAucuGAT: T 184 UcAGAUGAUGGAUuAAGAATsT AD-12139 336 ACAGUACACAACAAGGAUG 185 AcAGuAcAcAAcAAGGAuGT: T 186 cAUCCUUGUUGUGuACUGUTsT AD-121 4 0 337 AAGAAACUACGAUUGAUGG 187 AAGAAAcuAcGAuuGAuGGT: T 188 CcAUcAAUCGuAGUUUCUUTsT AD-121 4 1

EQ SEQ SEQ sequence of 19-mer antisense sequence (5 - duplex

[D ID sense sequence (5 -3 )) ID target site 3 ) name

0 : NO. NO.

338 AAACUACGAUUGAUGGAGA 189 AAAcuAcGAuuGAuGGAGAT: T 190 UCUCcAUcAAUCGuAGUUUTsT AD-121 4 2

339 UGGAGCUGUUGAUAAGAGA 191 uGGAGcuGuuGAUAAGAGAT: T 192 UCUCUuAUcAAcAGCUCcATsT AD-121 4 3

340 CUAACUAGAAUCCUCCAGG 193 cuAAcuAGAAuccuccAGGT: T 194 CCUGGAGGAUUCUAGUUAGTsT AD-12144

341 GAAUAUGCUCAUAGAGCAA 195 GAAuAuGCUCAuAGAGCAAT: T 196 UUGCUCuAUGAGcAuAUUCTsT AD-121 4 5

342 AUGCUCAUAGAGCAAAGAA 197 AuGCUCAuAGAGCAAAGAAT: T 198 UUCUUUGCUCuAUGAGcAUTsT AD-121 4 6

343 AAAAAUUGGUGCUGUUGAG 199 AAAAAuuGGuGcuGuuGAGT: T 200 CUcAAcAGcACcAAUUUUUTsT AD-121 4 7

344 GAGGAGCUGAAUAGGGUUA 201 GAGGAGcuGAAuAGGGuuAT: T 202 uAACCCuAUUcAGCUCCUCTsT AD-121 4 8

345 GGAGCUGAAUAGGGUUACA 203 GGAGcuGAAuAGGGuuAcAT: T 204 UGuAACCCuAUUcAGCUCCTsT AD-121 4 9

346 GAGCUGAAUAGGGUUACAG GAGcuGAAuAGGGuuAcAGT: T 206 CUGuAACCCuAUUcAGCUCTsT AD-12150

347 AGCUGAAUAGGGUUACAGA 207 AGcuGAAuAGGGuuAcAGAT: T 208 UCUGuAACCCuAUUcAGCUTsT AD-12151

348 GCUGAAUAGGGUUACAGAG 209 GcuGAAuAGGGuuAcAGAGT: T 210 CUCUGuAACCCuAUUcAGCTsT AD-12152

349 CCAAACUGGAUCGUAAGAA 211 CCAAAcuGGAucGuAAGAAT: T 212 UUCUuACGAUCcAGUUUGGTsT AD-12153

350 GAUCGUAAGAAGGCAGUUG 213 GAucGuAAGAAGGcAGuuGT: T 214 cAACUGCCUUCUuACGAUCTsT AD-1215 4

351 ACCUUAUUUGGUAAUCUGC 215 AccuuAuuuGGuAAucuGcT: T 216 GcAGAUuACcAAAuAAGGUTsT AD-12155

352 UUAGAUACCAUUACUACAG 217 uuAGAuAccAuuAcuAcAGT: T 218 CUGuAGuAAUGGuAUCuAATsT AD-12156

353 AUACCAUUACUACAGUAGC 219 AuAccAuuAcuAcAGuAGcT: T 220 GCuACUGuAGuAAUGGuAUTsT AD-12157

354 UACUACAGUAGCACUUGGA 221 uAcuAcAGuAGcAcuuGGAT: T 222 UCcAAGUGCuACUGuAGuATsT AD-12158

355 AAAGUAAAACUGUACUACA 223 AAAGuAAAAcuGuAcuAcAT: T 224 UGuAGuAcAGUUUuACUUUTsT AD-12159

356 CUCAAGACUGAUCUUCUAA 225 cucAAGAcuGAucuucuAAT: T 226 UuAGAAGAUcAGUCUUGAGTsT AD-12160

357 UUGACAGUGGCCGAUAAGA 227 uuGAcAGuGGccGAuAAGAT: T 228 UCUuAUCGGCcACUGUcAATsT AD-12161

358 UGACAGUGGCCGAUAAGAU 229 uGAcAGuGGccGAuAAGAuT: T 230 AUCUuAUCGGCcACUGUcATsT AD-12162

359 GCAAUGUGGAAACCUAACU 231 GcAAuGuGGAAAccuAAcuT: T 232 AGUuAGGUUUCcAcAUUGCTsT AD-12163

360 CCACUUAGUAGUGUCCAGG 233 ccAcuuAGuAGuGuccAGGT: T 234 CCUGGAcACuACuAAGUGGTsT AD-1216 4

361 AGAAGGUACAAAAUUGGUU 235 AGAAGGuAcAAAAuuGGuuT: T 236 AACcAAUUUUGuACCUUCUTsT AD-12165

362 UGGUUUGACUAAGCUUAAU 237 uGGuuuGAcuAAGcuuAAuT: T 238 AUuAAGCUuAGUcAAACcATsT AD-12166

363 GGUUUGACUAAGCUUAAUU 239 GGuuuGAcuAAGcuuAAuuT: T 240 AAUuAAGCUuAGUcAAACCTsT AD-12167

364 UCUAAGUCAAGAGCCAUCU 241 ucuAAGucAAGAGccAucuT: T 242 AGAUGGCUCUUGACUUAGATST AD-12168

365 UCAUCCCUAUAGUUCACUU 243 ucAucccuAuAGuucAcuuT: T 244 AAGUGAACuAuAGGGAUGATsT AD-12169

366 CAUCCCUAUAGUUCACUUU 245 cAucccuAuAGuucAcuuuT: T 246 AAAGUGAACuAuAGGGAUGTsT AD-12170

367 CCCUAGACUUCCCUAUUUC 247 cccuAGAcuucccuAuuucT: T 248 GAAAuAGGGAAGUCuAGGGTsT AD-12171

368 AGACUUCCCUAUUUCGCUU 249 AGAcuucccuAuuucGcuuT: T 250 AAGCGAAAuAGGGAAGUCUTsT AD-12172

369 UCACCAAACCAUUUGUAGA 251 ucAccAAAccAuuuGuAGAT: T 252 UCuAcAAAUGGUUUGGUGATsT AD-12173

370 UCCUUUAAGAGGCCUAACU 253 uccuuuAAGAGGccuAAcuT: T 254 AGUuAGGCCUCUuAAAGGATsT AD-1217 4

371 UUUAAGAGGCCUAACUCAU 255 uuuAAGAGGccuAAcucAuT: T 256 AUGAGUuAGGCCUCUuAAATsT AD-12175

372 UUAAGAGGCCUAACUCAUU 257 uuAAGAGGccuAAcucAuuT: T 258 AAUGAGUuAGGCCUCUuAATsT AD-12176

373 GGCCUAACUCAUUCACCCU 259 GGccuAAcucAuucAcccuT: T 260 AGGGUGAAUGAGUUAGGCCTST AD-12177

374 UGGUAUUUUUGAUCUGGCA 261 uGGuAuuuuuGAucuGGcAT: T 262 UGCcAGAUcAAAAAuACcATsT AD-12178

375 AGUUUAGUGUGUAAAGUUU 263 AGuuuAGuGuGuAAAGuuuT: T 264 AAACUUuAcAcACuAAACUTsT AD-12179

376 GCCAAAUUCGUCUGCGAAG 265 GccAAAuucGucuGcGAAGT: T 266 CUUCGcAGACGAAUUUGGCTsT AD-12180

377 AAUUCGUCUGCGAAGAAGA 267 AAuucGuCuGcGAAGAAGAT: T 268 UCUUCUUCGcAGACGAAUUTsT AD-12181

378 UGAAAGGUCACCUAAUGAA 269 uGAAAGGucAccuAAuGAAT: T 270 UUcAUuAGGUGACCUUUcATsT AD-12182

379 CAGACCAUUUAAUUUGGCA 271 cAGAccAuuuAAuuuGGcAT: T 272 UGCcAAAUuAAAUGGUCUGTsT AD-12183

380 AGACCAUUUAAUUUGGCAG 273 AGAccAuuuAAuuuGGcAGT: T 274 CUGCcAAAUuAAAUGGUCUTsT AD-1218 4

381 AGUUAUUAUGGGCUAUAAU 275 AGuuAuuAuGGGcuAuAAuT: T 276 AUuAuAGCCcAuAAuAACUTsT AD-12185

382 GCUGGUAUAAUUCCACGUA 277 GcuGGuAuAAuuccAcGuAT: T 278 uACGUGGAAUuAuACcAGCTsT AD-12186

383 AUUUAAUUUGGCAGAGCGG 279 AuuuAAuuuGGcAGAGcGGT: T 280 CCGCUCUGCcAAAUuAAAUTsT AD-12187

384 UUUAAUUUGGCAGAGCGGA 281 uuuAAuuuGGcAGAGcGGAT: T 282 UCCGCUCUGCcAAAUuAAATsT AD-12188

385 UUUGGCAGAGCGGAAAGCU 283 uuuGGcAGAGcGGAAAGcuT: T 284 AGCUUUCCGCUCUGCCAAATST AD-12189

386 UUUUACAAUGGAAGGUGAA 285 uuuuAcAAuGGAAGGuGAAT: T 286 UUcACCUUCcAUUGuAAAATsT AD-12190

387 AAUGGAAGGUGAAAGGUCA 287 AAuGGAAGGuGAAAGGuCAT: T 288 UGACCUUUcACCUUCcAUUTsT AD-12191

388 UGAGAUGCAGACCAUUUAA 289 uGAGAuGcAGAccAuuuAAT: T 290 UuAAAUGGUCUGcAUCUcATsT AD-12192

389 UCGCAGCCAAAUUCGUCUG 291 ucGcAGccAAAuucGucuGT: T 292 cAGACGAAUUUGGCUGCGATsT AD-12193

390 GGCUAUAAUUGCACUAUCU 293 GGcuAuAAuuGcAcuAucuT: T 294 AGAuAGUGcAAUuAuAGCCTsT AD-1219 4

391 AUUGACAGUGGCCGAUAAG 295 AuuGAcAGuGGccGAuAAGT: T 296 CUuAUCGGCcACUGUcAAUTsT AD-12195

392 CUAGACUUCCCUAUUUCGC 297 cuAGAcuucccuAuuucGcT: T 298 GCGAAAuAGGGAAGUCuAGTsT AD-12196

393 ACUAUCUUUGCGUAUGGCC 299 AcuAucuuuGcGuAuGGccT: T 300 GGCcAuACGcAAAGAuAGUTsT AD-12197

394 AUACUCUAGUCGUUCCCAC 301 AuAcucuAGucGuucccAcT: T 302 GUGGGAACGACUAGAGUAUTsT AD-12198

395 AAAGAAACUACGAUUGAUG 303 AAAGAAAcuAcGAuuGAuGT: T 304 cAUcAAUCGuAGUUUCUUUTsT AD-12199

396 GCCUUGAUUUUUUGGCGGG 305 GccuuGAuuuuuuGGcGGGT: T 306 CCCGCcAAAAAAUcAAGGCTsT AD-12200

397 CGCCCAUUCAAUAGUAGAA 307 CGCCCAuUcAAuAGuAGAAT: T 308 UUCuACuAUUGAAUGGGCGTsT AD-12201

398 CCUUAUUUGGUAAUCUGCU 309 ccuuAuuuGGuAAucuGcuT: T 310 AGcAGAUuACcAAAuAAGGTsT AD-12202

399 AGAGACAAUUCCGGAUGUG 311 AGAGAcAAuuCCGGAuGuGT: T 312 cAcAUCCGGAAUUGUCUCUTsT AD-12203

100 UGACUUUGAUAGCUAAAUU 313 uGAcuuuGAuAGcuAAAuuT: T 314 AAUUuAGCuAUcAAAGUcATsT AD-1220 4

101 UGGCAGAGCGGAAAGCUAG 315 uGGcAGAGcGGAAAGcuAGT: T 316 CuAGCUUUCCGCUCUGCcATsT AD-12205

502 GAGCGGAAAGCUAGCGCCC 317 GAGcGGAAAGcuAGcGcccT: T 318 GGGCGCuAGCUUUCCGCUCTsT AD-12206

503 AAAGAAGUUAGUGUACGAA 319 AAAGAAGuuAGuGuAcGAAT: T 320 UUCGuAcACuAACUUCUUUTsT AD-12207

504 AUUGCACUAUCUUUGCGUA 321 AuuGcAcuAucuuuGcGuAT: T 322 uACGcAAAGAuAGUGcAAUTsT AD-12208

505 GGUAUAAUUCCACGUACCC 323 GGuAuAAuuccAcGuAcccT: T 324 GGGuACGUGGAAUuAuACCTsT AD-12209

506 UACUCUAGUCGUUCCCACU 325 uAcucuAGucGuucccAcuT: T 326 AGUGGGAACGACUAGAGUATsT AD-12210

507 UAUGAAAGAAACUACGAUU 327 uAuGAAAGAAAcuAcGAuuT: T 328 AAUCGuAGUUUCUUUcAuATsT AD-12211

508 AUGCUAGAAGUACAUAAGA 329 AuGCUAGAAGuAcAuAAGAT: T 330 UCUuAUGuACUUCuAGcAUTsT AD-12212

509 AAGUACAUAAGACCUUAUU 331 AAGuAcAuAAGAcCUuAuuT: T 332 AAuAAGGUCUuAUGuACUUTsT AD-12213

EQ SEQ SEQ

[D sequence of 19-mer antisense sequence (5 - duplex

ID sense sequence (5 -3 )) ID

0 : target site NO. NO. 3 ) name

110 ACAGCCUGAGCUGUUAAUG 333 AcAGccuGAGcuGuuAAuGT: T 334 cAUuAAcAGCUcAGGCUGUTsT AD-1221 4

511 AAAGAAGAGACAAUUCCGG 335 AAAGAAGAGAcAAuuCCGGT: T 336 CCGGAAUUGUCUCUUCUUUTST AD-12215

512 CACACUGGAGAGGUCUAAA 337 cAcAcuGGAGAGGucuAAAT: T 338 UUuAGACCUCUCcAGUGUGTsT AD-12216

513 CACUGGAGAGGUCUAAAGU 339 cAcuGGAGAGGucuAAAGuT: T 340 ACUUuAGACCUCUCcAGUGTsT AD-12217

514 ACUGGAGAGGUCUAAAGUG 341 AcuGGAGAGGucuAAAGuGT: T 342 cACUUuAGACCUCUCcAGUTsT AD-12218

515 CGUCGCAGCCAAAUUCGUC 343 cGucGcAGccAAAuucGucT: T 344 GACGAAUUUGGCUGCGACGTST AD-12219

516 GAAGGCAGUUGACCAACAC 345 GAAGGcAGuuGAccAAcAcT: T 346 GUGUUGGUcAACUGCCUUCTsT AD-12220

517 CAUUCACCCUGACAGAGUU 347 cAuucAcccuGAcAGAGuuT: T 348 AACUCUGUcAGGGUGAAUGTsT AD-12221

518 AAGAGGCCUAACUCAUUCA 349 AAGAGGccuAAcucAuucAT: T 350 UGAAUGAGUuAGGCCUCUUTsT AD-12222

519 GAGACAAUUCCGGAUGUGG 351 GAGAcAAuuccGGAuGuGGT: T 352 CcAcAUCCGGAAUUGUCUCTsT AD-12223

520 UUCCGGAUGUGGAUGUAGA 353 uuccGGAuGuGGAuGuAGAT: T 354 UCuAcAUCcAcAUCCGGAATsT AD-1222 4

521 AAGCUAGCGCCCAUUCAAU 355 AAGcuAGcGcccAuucAAuT: T 356 AUUGAAUGGGCGCUAGCUUTST AD-12225

522 GAAGUUAGUGUACGAACUG 357 GAAGuuAGuGuAcGAAcuGT: T 358 cAGUUCGuAcACuAACUUCTsT AD-12226

523 UAUAAUUCCACGUACCCUU 359 uAuAAuuccAcGuAcccuuT: T 360 AAGGGuACGUGGAAUuAuATsT AD-12227

524 ACAGUGGCCGAUAAGAUAG 361 AcAGuGGccGAuAAGAuAGT: T 362 CuAUCUuAUCGGCcACUGUTsT AD-12228

525 UCUGUCAUCCCUAUAGUUC 363 ucuGucAucccuAuAGuucT: T 364 GAACuAuAGGGAUGAcAGATsT AD-12229

526 UUCUUGCUAUGACUUGUGU 365 uucuuGcuAuGAcuuGuGuT; T 366 AcAcAAGUcAuAGcAAGAATsT AD-12230

527 GUAAGAAGGCAGUUGACCA 367 GuAAGAAGGcAGuuGAccAT: T 368 UGGUcAACUGCCUUCUuACTsT AD-12231

528 CAUUGACAGUGGCCGAUAA 369 cAuuGAcAGuGGccGAuAAT: T 370 UuAUCGGCcACUGUcAAUGTsT AD-12232

529 AGAAACCACUUAGUAGUGU 371 AGAAAccAcuuAGuAGuGuT: T 372 AcACuACuAAGUGGUUUCUTsT AD-12233

530 GGAUUGUUCAUCAAUUGGC 373 GGAuuGuucAucAAuuGGcT: T 374 GCcAAUUGAUGAAcAAUCCTsT AD-1223 4

531 UAAGAGGCCUAACUCAUUC 375 uAAGAGGccuAAcucAuucT: T 376 GAAUGAGUuAGGCCUCUuATsT AD-12235

532 AGUUAGUGUACGAACUGGA 377 AGuuAGuGuAcGAAcuGGAT: T 378 UCcAGUUCGuAcACuAACUTsT AD-12236

533 AGUACAUAAGACCUUAUUU 379 AGuAcAuAAGAccuuAuuuT: T 380 AAAuAAGGUCUuAUGuACUTsT AD-12237

534 UGAGCCUUGUGUAUAGAUU 381 uGAGccuuGuGuAuAGAuuT: T 382 AAUCuAuAcAcAAGGCUcATsT AD-12238

535 CCUUUAAGAGGCCUAACUC 383 ccuuuAAGAGGccuAAcucT: T 384 GAGUuAGGCCUCUuAAAGGTsT AD-12239

536 ACCACUUAGUAGUGUCCAG 385 AccAcuuAGuAGuGuccAGT: T 386 CUGGAcACuACuAAGUGGUTsT AD-122 4 0

537 GAAACUUCCAAUUAUGUCU 387 GAAAcuuccAAuuAuGucuT: T 388 AGAcAuAAUUGGAAGUUUCTsT AD-122 4 1

538 UGCAUACUCUAGUCGUUCC 389 uGcAuAcucuAGucGuuccT: T 390 GGAACGACuAGAGuAUGcATsT AD-122 4 2

539 AGAAGGCAGUUGACCAACA 391 AGAAGGcAGuuGAccAAcAT: T 392 UGUUGGUcAACUGCCUUCUTsT AD-122 4 3

540 GUACAUAAGACCUUAUUUG 393 GuAcAuAAGAccuuAuuuGT: T 394 cAAAuAAGGUCUuAUGuACTsT AD-12244

541 UAUAAUUGCACUAUCUUUG 395 uAuAAuuGcAcuAucuuuGT: T 396 cAAAGAuAGUGcAAUuAuATsT AD-122 4 5

542 UCUCUGUUACAAUACAUAU 397 ucucuGuuAcAAuAcAuAuT: T 398 AuAUGuAUUGuAAcAGAGATsT AD-122 4 6

543 UAUGCUCAUAGAGCAAAGA 399 uAuGcucAuAGAGcAAAGAT: T 400 UCUUUGCUCuAUGAGcAuATsT AD-122 4 7

544 UGUUGUUUGUCCAAUUCUG 401 uGuuGuuuGuccAAuucuGT: T 402 cAGAAUUGGAcAAAcAAcATsT AD-122 4 8

545 ACUAACUAGAAUCCUCCAG 403 AcuAAcuAGAAuccuccAGT: T 404 CUGGAGGAUUCUAGUUAGUTsT AD-122 4 9

546 UGUGGUGUCUAUACUGAAA 405 uGuGGuGucuAuAcuGAAAT: T 406 UUUcAGuAuAGAcACcAcATsT AD-12250

547 UAUUAUGGGAGACCACCCA 407 uAuuAuGGGAGAccAcccAT: T 408 UGGGUGGUCUCCcAuAAuATsT AD-12251

548 AAGGAUGAAGUCUAUCAAA 409 AAGGAuGAAGuCuAucAAAT: T 410 UUUGAuAGACUUcAUCCUUTsT AD-12252

549 UUGAUAAGAGAGCUCGGGA 411 uuGAuAAGAGAGcucGGGAT: T 412 UCCCGAGCUCUCUUAUCAATsT AD-12253

550 AUGUUCCUUAUCGAGAAUC 413 AuGuuccuuAucGAGAAucT: T 414 GAUUCUCGAuAAGGAAcAUTsT AD-1225 4

551 GGAAUAUGCUCAUAGAGCA 415 GGAAuAuGcucAuAGAGcAT: T 416 UGCUCuAUGAGcAuAUUCCTsT AD-12255

552 CCAUUCCAAACUGGAUCGU 417 ccAuuccAAAcuGGAucGuT: T 418 ACGAUCcAGUUUGGAAUGGTsT AD-12256

553 GGCAGUUGACCAACACAAU 419 GGcAGuuGAccAAcAcAAuT: T 420 AUUGUGUUGGUcAACUGCCTsT AD-12257

554 CAUGCUAGAAGUACAUAAG 421 cAuGcuAGAAGuAcAuAAGT: T 422 CUuAUGuACUUCuAGcAUGTsT AD-12258

555 CUAGAAGUACAUAAGACCU 423 cuAGAAGuAcAuAAGAccuT: T 424 AGGUCUuAUGuACUUCuAGTsT AD-12259

556 UUGGAUCUCUCACAUCUAU 425 uuGGAucucucAcAucuAuT: T 426 AuAGAUGUGAGAGAUCcAATsT AD-12260

557 AACUGUGGUGUCUAUACUG 427 AAcuGuGGuGucuAuAcuGT: T 428 cAGuAuAGAcACcAcAGUUTsT AD-12261

558 UCAUUGACAGUGGCCGAUA 429 ucAuuGAcAGuGGccGAuAT: T 430 uAUCGGCcACUGUcAAUGATsT AD-12262

559 AUAAAGCAGACCCAUUCCC 431 AuAAAGcAGAcccAuucccT: T 432 GGGAAUGGGUCUGCUUUAUTST AD-12263

560 ACAGAAACCACUUAGUAGU 433 AcAGAAAccAcuuAGuAGuT: T 434 ACuACuAAGUGGUUUCUGUTsT AD-1226 4

561 GAAACCACUUAGUAGUGUC 435 GAAAccAcuuAGuAGuGucT: T 436 GAcACuACuAAGUGGUUUCTsT AD-12265

562 AAAUCUAAGGAUAUAGUCA 437 AAAuCuAAGGAuAuAGucAT: T 438 UGACuAuAUCCUuAGAUUUTsT AD-12266

563 UUAUUUAUACCCAUCAACA 439 uuAuuuAuAcccAucAAcAT: T 440 UGUUGAUGGGuAuAAAuAATsT AD-12267

564 ACAGAGGCAUUAACACACU 441 AcAGAGGcAuuAAcAcAcuT: T 442 AGUGUGUuAAUGCCUCUGUTsT AD-12268

565 ACACACUGGAGAGGUCUAA 443 AcAcAcuGGAGAGGuCuAAT: T 444 UuAGACCUCUCcAGUGUGUTsT AD-12269

566 ACACUGGAGAGGUCUAAAG 445 AcAcuGGAGAGGucuAAAGT: T 446 CUUuAGACCUCUCcAGUGUTsT AD-12270

567 CGAGCCCAGAUCAACCUUU 447 cGAGcccAGAucAAccuuuT: T 448 AAAGGUUGAUCUGGGCUCGTST AD-12271

568 UCCCUAUUUCGCUUUCUCC 449 ucccuAuuucGcuuucuccT: T 450 GGAGAAAGCGAAAUAGGGATST AD-12272

569 UCUAAAAUCACUGUCAACA 451 uCuAAAAucAcuGucAAcAT: T 452 UGUUGAcAGUGAUUUuAGATsT AD-12273

570 AGCCAAAUUCGUCUGCGAA 453 AGccAAAuucGucuGcGAAT: T 454 UUCGcAGACGAAUUUGGCUTsT AD-1227 4

571 CCCAUUCAAUAGUAGAAUG 455 CCCAuucAAuAGuAGAAuGT: T 456 cAUUCuACuAUUGAAUGGGTsT AD-12275

572 GAUGAAUGCAUACUCUAGU 457 GAuGAAuGcAuAcucuAGuT: T 458 ACuAGAGuAUGcAUUcAUCTsT AD-12276

573 CUCAUGUUCCUUAUCGAGA 459 cucAuGuuccuuAucGAGAT: T 460 UCUCGAuAAGGAAcAUGAGTsT AD-12277

574 GAGAAUCUAAACUAACUAG 461 GAGAAuCuAAAcuAAcuAGT: T 462 CuAGUuAGUUuAGAUUCUCTsT AD-12278

575 UAGAAGUACAUAAGACCUU 463 uAGAAGuAcAuAAGAccuuT: T 464 AAGGUCUuAUGuACUUCuATsT AD-12279

576 CAGCCUGAGCUGUUAAUGA 465 cAGccuGAGcuGuuAAuGAT: T 466 UcAUuAAcAGCUcAGGCUGTsT AD-12280

577 AAGAAGAGACAAUUCCGGA 467 AAGAAGAGAcAAuuCCGGAT: T 468 UCCGGAAUUGUCUCUUCUUTST AD-12281

578 UGCUGGUGUGGAUUGUUCA 469 uGcuGGuGuGGAuuGuucAT: T 470 UGAAcAAUCcAcACcAGcATsT AD-12282

579 AAAUUCGUCUGCGAAGAAG 471 AAAuucGucuGcGAAGAAGT: T 472 CUUCUUCGcAGACGAAUUUTsT AD-12283

580 UUUCUGGAAGUUGAGAUGU 473 uuucuGGAAGuuGAGAuGuT: T 474 AcAUCUcAACUUCcAGAAATsT AD-1228 4

581 UACUAAACAGAUUGAUGUU 475 uAcuAAAcAGAuuGAuGuuT: T 476 AAcAUcAAUCUGUUuAGuATsT AD-12285

EQ SEQ SEQ sequence of 19-mer antisense sequence (5 - duplex

[D ID sense sequence (5 -3 )) ID target site 3 ) name

0 : NO. NO.

582 GAUUGAUGUUUACCGAAGU 477 GAuuGAuGuuuAccGAAGuT: T 478 ACUUCGGuAAAcAUcAAUCTsT AD-12286

583 GCACUAUCUUUGCGUAUGG 479 GcAcuAucuuuGcGuAuGGT: T 480 CcAuACGcAAAGAuAGUGCTsT AD-12287

584 UGGUAUAAUUCCACGUACC 481 uGGuAuAAuuccAcGuAccT: T 482 GGuACGUGGAAUuAuACcATsT AD-12288

585 AGCAAGCUGCUUAACACAG 483 AGcAAGcuGcuuAAcAcAGT: T 484 CUGUGUuAAGcAGCUUGCUTsT AD-12289

586 CAGAAACCACUUAGUAGUG 485 cAGAAAccAcuuAGuAGuGT: T 486 cACuACuAAGUGGUUUCUGTsT AD-12290

587 AACUUAUUGGAGGUUGUAA 487 AAcuuAuuGGAGGuuGuAAT: T 488 UuAcAACCUCcAAuAAGUUTsT AD-12291

588 CUGGAGAGGUCUAAAGUGG 489 cuGGAGAGGucuAAAGuGGT: T 490 CcACUUuAGACCUCUCcAGTsT AD-12292

589 AAAAAAGAUAUAAGGCAGU 491 AAAAAAGAuAuAAGGcAGuT: T 492 ACUGCCUuAuAUCUUUUUUTsT AD-12293

590 GAAUUUUGAUAUCUACCCA 493 GAAuuuuGAuAucuAcccAT: T 494 UGGGuAGAuAUcAAAAUUCTsT AD-1229 4

591 GUAUUUUUGAUCUGGCAAC 495 GuAuuuuuGAucuGGcAAcT: T 496 GUUGCcAGAUcAAAAAuACTsT AD-12295

592 AGGAUCCCUUGGCUGGUAU 497 AGGAucccuuGGcuGGuAuT: T 498 AuACcAGCcAAGGGAUCCUTsT AD-12296

593 GGAUCCCUUGGCUGGUAUA 499 GGAucccuuGGcuGGuAuAT: T 500 uAuACcAGCcAAGGGAUCCTsT AD-12297

594 CAAUAGUAGAAUGUGAUCC 501 cAAuAGuAGAAuGuGAuCCT: T 502 GGAUcAcAUUCuACuAUUGTsT AD-12298

595 GCUAUAAUUGCACUAUCUU 503 GcuAuAAuuGcAcuAucuuT: T 504 AAGAuAGUGcAAUuAuAGCTsT AD-12299

596 UACCCUUCAUCAAAUUUUU 505 uAcccuucAucAAAuuuuuT: T 506 AAAAAUUUGAUGAAGGGUATST AD-12300

597 AGAACAUAUUGAAUAAGCC 507 AGAAcAuAuuGAAuAAGccT: T 508 GGCUuAUUcAAuAUGUUCUTsT AD-12301

598 AAAUUGGUGCUGUUGAGGA 509 AAAuuGGuGcuGuuGAGGAT: T 510 UCCUcAAcAGcACcAAUUUTsT AD-12302

599 UGAAUAGGGUUACAGAGUU 511 uGAAuAGGGuuAcAGAGuuT: T 512 AACUCUGuAACCCuAUUcATsT AD-12303

300 AAGAACUUGAAACCACUCA 513 AAGAAcuuGAAAccAcucAT: T 514 UGAGUGGUUUcAAGUUCUUTsT AD-1230 4

DOI AAUAAAGCAGACCCAUUCC 515 AAuAAAGcAGAcccAuuCCT: T 516 GGAAUGGGUCUGCUUUAUUTST AD-12305 AUACCCAUCAACACUGGUA 517 AuAcccAucAAcAcuGGuAT: T 518 uACcAGUGUUGAUGGGuAUTsT AD-12306

DO3 UGGAUUGUUCAUCAAUUGG 519 uGGAuuGuucAucAAuuGGT: T 520 CcAAUUGAUGAAcAAUCcATsT AD-12307 304 UGGAGAGGUCUAAAGUGGA 521 uGGAGAGGucuAAAGuGGAT: T 522 UCcACUUuAGACCUCUCcATsT AD-12308 305 GUCAUCCCUAUAGUUCACU 523 GucAucccuAuAGuucAcuT: T 524 AGUGAACuAuAGGGAUGACTsT AD-12309 306 AUAAUGGCUAUAAUUUCUC 525 AuAAuGGcuAuAAuuucucT: T 526 GAGAAAUuAuAGCcAUuAUTsT AD-12310 307 AUCCCUUGGCUGGUAUAAU 527 AucccuuGGcuGGuAuAAuT: T 528 AUuAuACcAGCcAAGGGAUTsT AD-12311 308 GGGCUAUAAUUGCACUAUC 529 GGGcuAuAAuuGcAcuAucT: T 530 GAuAGUGcAAUuAuAGCCCTsT AD-12312 309 GAUUCUCUUGGAGGGCGUA 531 GAuucucuuGGAGGGcGuAT: T 532 uACGCCCUCcAAGAGAAUCTsT AD-12313

DlO GCAUCUCUCAAUCUUGAGG 533 GcAucucucAAucuuGAGGT: T 534 CCUcAAGAUUGAGAGAUGCTsT AD-1231 4 DlI CAGCAGAAAUCUAAGGAUA 535 cAGcAGAAAuCuAAGGAuAT: T 536 uAUCCUuAGAUUUCUGCUGTsT AD-12315

D12 GUCAAGAGCCAUCUGUAGA 537 GucAAGAGccAucuGuAGAT: T 538 UCuAcAGAUGGCUCUUGACTsT AD-12316 D13 AAACAGAGGCAUUAACACA 539 AAAcAGAGGcAuuAAcAcAT: T 540 UGUGUuAAUGCCUCUGUUUTsT AD-12317 D14 AGCCCAGAUCAACCUUUAA 541 AGcccAGAucAAccuuuAAT: T 542 UuAAAGGUUGAUCUGGGCUTsT AD-12318 D15 UAUUUUUGAUCUGGCAACC 543 uAuuuuuGAucuGGcAAccT: T 544 GGUUGCcAGAUcAAAAAuATsT AD-12319 D16 UGUUUGGAGCAUCUACUAA 545 uGuuuGGAGcAucuAcuAAT: T 546 UuAGuAGAUGCUCcAAAcATsT AD-12320 D17 GAAAUUACAGUACACAACA 547 GAAAuuAcAGuAcAcAAcAT: T 548 UGUUGUGuACUGuAAUUUCTsT AD-12321 D18 ACUUGACCAGUGUAAAUCU 549 AcuuGAccAGuGuAAAucuT: T 550 AGAUUuAcACUGGUcAAGUTsT AD-12322 D19 ACCAGUGUAAAUCUGACCU 551 AccAGuGuAAAucuGAccuT: T 552 AGGUcAGAUUuAcACUGGUTsT AD-12323 D20 AGAACAAUCAUUAGCAGCA 553 AGAAcAAucAuuAGcAGcAT: T 554 UGCUGCuAAUGAUUGUUCUTsT AD-1232 4 D21 CAAUGUGGAAACCUAACUG 555 cAAuGuGGAAAccuAAcuGT: T 556 cAGUuAGGUUUCcAcAUUGTsT AD-12325 D22 ACCAAGAAGGUACAAAAUU 557 AccAAGAAGGuAcAAAAuuT: T 558 AAUUUUGuACCUUCUUGGUTsT AD-12326 D23 GGUACAAAAUUGGUUGAAG 559 GGuAcAAAAuuGGuuGAAGT: T 560 CUUcAACcAAUUUUGuACCTsT AD-12327 D24 GGUGUGGAUUGUUCAUCAA 561 GGuGuGGAuuGuucAucAAT: T 562 UUGAUGAAcAAUCcAcACCTsT AD-12328 D25 AGAGUUCACAAAAAGCCCA 563 AGAGuucAcAAAAAGcccAT: T 564 UGGGCUUUUUGUGAACUCUTST AD-12329 D26 UGAUAGCUAAAUUAAACCA 565 uGAuAGcuAAAuuAAAccAT: T 566 UGGUUuAAUUuAGCuAUcATsT AD-12330 D27 AAUAAGCCUGAAGUGAAUC 567 AAuAAGccuGAAGuGAAucT: T 568 GAUUcACUUcAGGCUuAUUTsT AD-12331 D28 CAGUUGACCAACACAAUGC 569 cAGuuGAccAAcAcAAuGcT: T 570 GcAUUGUGUUGGUcAACUGTsT AD-12332 D29 UGGUGUGGAUUGUUCAUCA 571 uGGuGuGGAuuGuucAucAT: T 572 UGAUGAAcAAUCcAcACCATsT AD-12333 D30 AUUCACCCUGACAGAGUUC 573 AuucAcccuGAcAGAGuucT: T 574 GAACUCUGUcAGGGUGAAUTsT AD-1233 4 D31 UAAGACCUUAUUUGGUAAU 575 uAAGAccuuAuuuGGuAAuT: T 576 AUuACcAAAuAAGGUCUuATsT AD-12335 D32 AAGCAAUGUGGAAACCUAA 577 AAGcAAuGuGGAAAccuAAT: T 578 UuAGGUUUCcAcAUUGCUUTsT AD-12336 D33 UCUGAAACUGGAUAUCCCA 579 ucuGAAAcuGGAuAucccAT: T 580 UGGGAuAUCcAGUUUcAGATsT AD-12337

Table 2b. Analysis of Eg5/KSP dsRNA duplexes

Table 3. Sequences and analysis of Eg5/KSP dsRNA duplexes single SDs 2nd

SEQ SEQ dose

Antisense sequence (5 screen @ screen

Sense sequence (5 -3 ) ID ID duplex (among 3 ) 25 nM [ %

NO. NO . name residual quadru mRNA] plicat es ) ccAuuAcuAcAGuAGcAcuTsT 582 AGUGCuACUGuAGuAAUGGT 583 AD-1 4 085 19% 1 %

AucuGGcAAccAuAuuucuTsT 584 AGAAAUAUGGUUGCCAGAUT; 3T 585 AD-1 4 086 38 % 1 %

GAuAGcuAAAuuAAAccAATsT 586 UUGGUUuAAUUuAGCuAUCT; 3T 587 AD-1 4 087 75% 10 %

AGAuAccAuuAcuAcAGuATsT 588 uACUGuAGuAAUGGuAUCUT; 3T 589 AD-1 4 088 22 % 8 %

GAuuGuucAucAAuuGGcGTsT 590 CGCcAAUUGAUGAAcAAUCT; 3T 591 AD-1 4 089 70 % 12 %

GcuuucuccucGGcucAcuTsT 592 AGuGAGCCGAGGAGAAAGCT 3T 593 AD-1 4 090 79% 11 %

GGAGGAuuGGcuGAcAAGATsT 594 UCUUGUcAGCcAAUCCUCCT; 3T 595 AD-1 4 091 29% 3% uAAuGAAGAGuAuAccuGGTsT 596 CcAGGuAuACUCUUcAUuAT; 3T 597 AD-1 4 092 23% 2 % uuucAccAAAccAuuuGuATsT 598 uAcAAAUGGUUUGGUGAAAT; 3T 599 AD-1 4 093 60 % 2 %

CUuAuuAAGGAGuAuAcGGTsT 600 CCGuAuACUCCUuAAuAAGT; 3T 601 AD-1 4 09 4 11 % 3%

GAAAucAGAuGGAcGuAAGTsT 602 CUuACGUCcAUCUGAUUUCT; 3T 603 AD-1 4 095 10 % 2 % cAGAuGucAGcAuAAGcGATsT 604 UCGCUuAUGCUGAcAUCUGT; 3T 605 AD-1 4 096 27 % 2 %

AucuAAcccuAGuuGuAucTsT 606 GAuAcAACuAGGGUuAGAUT; 3T 607 AD-1 4 097 45% 6%

AAGAGcuuGuuAAAAucGGTsT 608 CCGAUUUuAAcAAGCUCUUT; 3T 609 AD-1 4 098 50 % 10 % uuAAGGAGuAuAcGGAGGATsT 610 UCCUCCGuAuACUCCUuAAT; 3T 611 AD-1 4 099 12 % 4 % uuGcAAuGuAAAuAcGuAuTsT 612 AuACGuAUUuAcAUUGcAAT; 3T 613 AD-1 4 100 49% 7 % ucuAAcccuAGuuGuAuccTsT 614 GGAuAcAACuAGGGUuAGAT; 3T 615 AD-1 4 101 36% 1 % cAuGuAucuuuuucucGAuTsT 616 AUCGAGAAAAAGAUACAUGT; 3T 617 AD-1 4 102 49% 3%

GAuGucAGcAuAAGcGAuGTsT 618 cAUCGCUuAUGCUGAcAUCT; 3T 619 AD-1 4 103 74 % 5% ucccAAcAGGuAcGAcAccTsT 620 GGUGUCGuACCUGUUGGGAT 3T 621 AD-I 4 IO 4 27 % 3% uGcucAcGAuGAGuuuAGuTsT 622 ACuAAACUcAUCGUGAGcAT 3T 623 AD-1 4 105 34 % 4 %

AGAGcuuGuuAAAAucGGATsT 624 UCCGAUUUuAAcAAGCUCUT 3T 625 AD-1 4 106 9% 2 %

GcGuAcAAGAAcAuCuAuATsT 626 uAuAGAUGUUCUUGuACGCT 3T 627 AD-1 4 107 5% 1 %

GAGGuuGuAAGccAAuGuuTsT 628 AAcAUUGGCUuAcAACCUCT 3T 629 AD-1 4 108 15% 1 %

AAcAGGuAcGAcAccAcAGTsT 630 CUGUGGUGUCGUACCUGUUT 3T 631 AD-1 4 109 91 % 2 %

AAcccuAGuuGuAucccucTsT 632 GAGGGAuAcAACuAGGGUUT 3T 633 AD-1 4 110 66% 5%

GcAuAAGcGAuGGAuAAuATsT 634 uAUuAUCcAUCGCUuAUGCT 3T 635 AD-1 4 111 33% 3%

AAGcGAuGGAuAAuAccuATsT 636 uAGGuAUuAUCcAUCGCUUT 3T 637 AD-1 4 112 51 % 3% uGAuCCuGuAcGAAAAGAATsT 638 UUCUUUUCGUACAGGAUCAT 3T 639 AD-1 4 113 22 % 3%

AAAAcAuuGGccGuucuGGTsT 640 CcAGAACGGCcAAUGUUUUT 3T 641 AD-I 4 H 4 117 % 8 % cuuGGAGGGcGuAcAAGAATsT 642 UUCUUGuACGCCCUCcAAGT 3T 643 AD-1 4 115 50 % 8 %

GGcGuAcAAGAAcAuCuAuTsT 644 AuAGAUGUUCUUGuACGCCT 3T 645 AD-1 4 116 14 % 3%

AcuCuGAGuAcAuuGGAAuTsT 646 AUUCcAAUGuACUcAGAGUT 3T 647 AD-1 4 117 12 % 4 % uuAuuAAGGAGuAuAcGGATsT 648 UCCGuAuACUCCUuAAuAAT 3T 649 AD-1 4 118 26% 4 % uAAGGAGuAuAcGGAGGAGTsT 650 CUCCUCCGuAuACUCCUuAT; 3T 651 AD-1 4 119 24 % 5%

AAAucAAuAGucAAcuAAATsT 652 UUuAGUUGACuAUUGAUUUT; 3T 653 AD-1 4 120 8 % 1 %

AAucAAuAGucAAcuAAAGTsT 654 CUUuAGUUGACuAUUGAUUT; 3T 655 AD-1 4 121 24 % 2 % uucucAGuAuAcuGuGuAATsT 656 UuAcAcAGuAuACUGAGAAT; 3T 657 AD-1 4 122 10 % 1 % uGuGAAAcAcuCuGAuAAATsT 658 UUuAUcAGAGUGUUUcAcAT; 3T 659 AD-1 4 123 8 % 1 %

AGAuGuGAAuCUCuGAAcATsT 660 UGUUcAGAGAUUcAcAUCUT; 3T 661 AD-1 4 12 4 9% 2 %

AGGuuGuAAGccAAuGuuGTsT 662 cAAcAUUGGCUuAcAACCUT; 3T 663 AD-1 4 125 114 % 6% uGAGAAAucAGAuGGAcGuTsT 664 ACGUCcAUCUGAUUUCUcAT; 3T 665 AD-1 4 126 9% 1 %

AGAAAucAGAuGGAcGuAATsT 666 UuACGUCcAUCUGAUUUCUT; 3T 667 AD-1 4 127 57 % 6%

AuAuCCCAAcAGGuAcGAcTsT 668 GUCGuACCUGUUGGGAuAUT; 3T 669 AD-1 4 128 104 % 6%

CCCAAcAGGuAcGAcAccATsT 670 UGGUGUCGuACCUGUUGGGT; 3T 671 AD-1 4 129 21 % 2 %

AGuAuAcuGAAGAAccuCuTsT 672 AGAGGUUCUUcAGuAuACUT; 3T 673 AD-1 4 130 57 % 6%

AuAuAuAucAGccGGGcGcTsT 674 GCGCCCGGCUGAUAUAUAUT; 3T 675 AD-1 4 131 93% 6%

AAucuAAcccuAGuuGuAuTsT 676 AuAcAACuAGGGUuAGAUUT; 3T 677 AD-1 4 132 75% 8 % cuAAcccuAGuuGuAucccTsT 678 GGGAuAcAACuAGGGUuAGT; 3T 679 AD-1 4 133 66% 4 % cuAGuuGuAucccuccuuuTsT 680 AAAGGAGGGAuAcAACuAGT; 3T 681 AD-1 4 13 4 44 % 6%

AGAcAucuGAcuAAuGGcuTsT 682 AGCcAUuAGUcAGAUGUCUT 3T 683 AD-1 4 135 55% 6%

GAAGcucAcAAuGAuuuAATsT 684 UuAAAUCAUUGUGAGCUUCT 3T 685 AD-1 4 136 29% 3%

AcAuGuAucuuuuucucGATsT 686 UCGAGAAAAAGAUACAUGUT 3T 687 AD-1 4 137 40 % 3% ucGAuucAAAucuuAAcccTsT 688 GGGUuAAGAUUUGAAUCGAT 3T 689 AD-1 4 138 39% 5%

SDs single 2nd dose

SEQ SEQ screen

Antisense sequence (5 duplex screen @

Sense sequence (5 -3 )) I D ID (among 3 ) name 25 nM [ %

NO . NO . quadru residual plicat mRNA] es ) ucuuAAcccuuAGGAcucuT; 3T 690 AGAGUCCuAAGGGUuAAGAT; 3T 691 AD-1 4 139 71 % 11 %

GcucAcGAuGAGuuuAGuGT; 3T 692 cACuAAACUcAUCGUGAGCT; 3T 693 AD-I 4 I 4 O 43% 15% cAuAAGcGAuGGAuAAuAcT; 3T 694 GuAUuAUCcAUCGCUuAUGT; 3T 695 AD-1 4 1 4 1 33% 6%

AuAAGCGAuGGAuAAuAcCT; 3T 696 GGuAUuAUCcAUCGCUuAUT; 3T 697 AD-1 4 1 4 2 51 % 14 % ccuAAuAAAcuGcccucAGT; 3T 698 CUGAGGGcAGUUuAUuAGGT; 3T 699 AD-1 4 1 4 3 42 % 1 % ucGGAAAGuuGAAcuuGGuT; 3T 700 ACcAAGUUCAACUUUCCGAT; 3T 701 AD-I 4 I 44 4 % 4 %

GAAAAcAuuGGccGuucuGT; 3T 702 cAGAACGGCcAAUGUUUUCT; 3T 703 AD-1 4 1 4 5 92 % 5%

AAGAcuGAuCUUCuAAGuuT 3T 704 AACUuAGAAGAUCAGUCUUT 3T 705 AD-1 4 1 4 6 13% 2 %

GAGcuuGuuAAAAucGGAAT 3T 706 UUCCGAUUUuAAcAAGCUCT 3T 707 AD-I 4 W 8 % 1 %

AcAuuGGccGuucuGGAGcT 3T 708 GCUCcAGAACGGCcAAUGUT 3T 709 AD-1 4 1 4 8 80 % 7 %

AAGAAcAuCuAuAAuuGcAT 3T 710 UGcAAUuAuAGAUGUUCUUT 3T 711 AD-1 4 1 4 9 44 % 7 %

AAAuGuGuCuAcucAuGuuT 3T 712 AAcAUGAGuAGAcAcAUUUT 3T 713 AD-1 4 150 32 % 29% uGucuAcucAuGuuucucAT 3T 714 UGAGAAAcAUGAGuAGAcAT 3T 715 AD-1 4 151 75% 11 %

GuAuAcuGuGuAAcAAuCuT 3T 716 AGAUUGUuAcAcAGuAuACT 3T 717 AD-1 4 152 8 % 5% uAuAcuGuGuAAcAAuCuAT 3T 718 uAGAUUGUuAcAcAGuAuAT 3T 719 AD-1 4 153 17 % 11 %

CUuAGuAGuGuCCAGGAAAT 3T 720 UUUCCUGGAcACuACuAAGT 3T 721 AD-1 4 15 4 16% 4 % ucAGAuGGAcGuAAGGcAGT 3T 722 CUGCCUuACGUCcAUCUGAT 3T 723 AD-1 4 155 11 % 1 %

AGAuAAAuuGAuAGcAcAAT 3T 724 UUGUGCuAUcAAUUuAUCUT 3T 725 AD-1 4 156 10 % 1 % cAAcAGGuAcGAcAccAcAT 3T 726 UGUGGUGUCGuACCUGUUGT 3T 727 AD-1 4 157 29% 3% uGcAAuGuAAAuAcGuAuuT 3T 728 AAuACGuAUUuAcAUUGcAT 3T 729 AD-1 4 158 51 % 3%

AGucAGAAuuuuAuCuAGAT 3T 730 UCuAGAuAAAAUUCUGACUT 3T 731 AD-1 4 159 53% 5%

CuAGAAAucuuuuAAcAcCT 3T 732 GGUGUuAAAAGAUUUCuAGT 3T 733 AD-1 4 160 40 % 3%

AAuAAAucuAAcCCuAGuuT 3T 734 AACuAGGGUuAGAUUuAUUT 3T 735 AD-1 4 161 83% 7 %

AAuuuucuGcucAcGAuGAT; 3T 736 UcAUCGUGAGcAGAAAAUUT; 3T 737 AD-1 4 162 44 % 6%

GcccucAGuAAAuccAuGGT; 3T 738 CcAUGGAUUuACUGAGGGCT; 3T 739 AD-1 4 163 57 % 3%

AcGuuuAAAAcGAGAuCUuT; 3T 740 AAGAUCUCGUUUUAAACGUT; 3T 741 AD-1 4 16 4 4 % 1 %

AGGAGAuAGAAcGuuuAAAT; 3T 742 UUuAAACGUUCuAUCUCCUT; 3T 743 AD-1 4 165 11 % 1 %

GAccGucAuGGcGucGcAGT; 3T 744 CUGCGACGCcAUGACGGUCT; 3T 745 AD-1 4 166 90 % 5%

AccGucAuGGcGucGcAGcT; 3T 746 GCUGCGACGCcAUGACGGUT; 3T 747 AD-1 4 167 49% 1 %

GAAcGuuuAAAAcGAGAucT; 3T 748 GAUCUCGUUUuAAACGUUCT; 3T 749 AD-1 4 168 12 % 2 % uuGAGcuuAAcAuAGGuAAT; 3T 750 UuACCuAUGUuAAGCUcAAT; 3T 751 AD-1 4 169 66% 4 %

AcuAAAuuGAuCUCGuAGAT; 3T 752 UCuACGAGAUcAAUUuAGUT; 3T 753 AD-1 4 170 52 % 6%

UcGuAGAAuuAucuuAAuAT; 3T 754 uAUuAAGAuAAUUCuACGAT; 3T 755 AD-1 4 171 42 % 4 %

GGAGAuAGAAcGuuuAAAAT; 3T 756 UUUuAAACGUUCuAUCUCCT; 3T 757 AD-1 4 172 3% 1 %

AcAAcuuAuuGGAGGuuGuT; 3T 758 AcAACCUCcAAuAAGUUGUT; 3T 759 AD-1 4 173 29% 2 % uGAGcuuAAcAuAGGuAAAT; 3T 760 UUUACCUAUGUuAAGCUCAT; 3T 761 AD-1 4 17 4 69% 2 %

AucucGuAGAAuuAucuuAT; 3T 762 uAAGAuAAUUCuACGAGAUT; 3T 763 AD-1 4 175 53% 3% cuGcGuGcAGucGGuccucT; 3T 764 GAGGACCGACUGCACGCAGT; 3T 765 AD-1 4 176 111 % 4 % cAcGcAGcGcccGAGAGuAT; 3T 766 uACUCUCGGGCGCUGCGUGT; 3T 767 AD-1 4 177 87 % 6%

AGuAccAGGGAGAcuccGGT 3T 768 CCGGAGUCUCCCUGGUACUT 3T 769 AD-1 4 178 59% 2 %

AcGGAGGAGAuAGAAcGuuT 3T 770 AACGUUCuAUCUCCUCCGUT 3T 771 AD-1 4 179 9% 2 %

AGAAcGuuuAAAAcGAGAuT 3T 772 AUCUCGUUUuAAACGUUCUT 3T 773 AD-1 4 180 43% 2 %

AAcGuuuAAAAcGAGAuCuT 3T 774 AGAUCUCGUUUUAAACGUUT 3T 775 AD-1 4 181 70 % 10 %

AGcuuGAGcuuAAcAuAGGT 3T 776 CCuAUGUuAAGCUcAAGCUT 3T 111 AD-1 4 182 100 % 7 %

AGcuuAAcAuAGGuAAAuAT 3T 778 uAUUuACCuAUGUuAAGCUT 3T 119 AD-1 4 183 60 % 5% uAGAGcuAcAAAAccuAucT 3T 780 GAuAGGUUUUGuAGCUCuAT 3T 781 AD-1 4 18 4 129% 6% uAGuuGuAucccuccuuuAT 3T 782 uAAAGGAGGGAuAcAACuAT 3T 783 AD-1 4 185 62 % 4 %

AccAcCcAGAcAuCuGAcuT 3T 784 AGUcAGAUGUCUGGGUGGUT 3T 785 AD-1 4 186 42 % 3%

AGAAAcuAAAuuGAuCUCGT 3T 786 CGAGAUcAAUUuAGUUUCUT 3T 787 AD-1 4 187 123% 12 %

UCUCGuAGAAuuAucuuAAT 3T 788 UuAAGAuAAUUCuACGAGAT 3T 789 AD-1 4 188 38 % 2 % cAAcuuAuuGGAGGuuGuAT 3T 790 uAcAACCUCcAAuAAGUUGT 3T 791 AD-1 4 189 13% 1 % uuGuAucccuccuuuAAGuT 3T 792 ACUuAAAGGAGGGAuAcAAT 3T 793 AD-1 4 190 59% 3% ucAcAAcuuAuuGGAGGuuT 3T 794 AACCUCcAAuAAGUUGUGAT 3T 795 AD-1 4 191 93% 3%

AGAAcuGuAcuCUUCUcAGT 3T 796 CUGAGAAGAGuAcAGUUCUT 3T 797 AD-1 4 192 45% 5%

GAGcuuAAcAuAGGuAAAuT; 3T 798 AUUuACCuAUGUuAAGCUCT; 3T 799 AD-1 4 193 57 % 3% cAccAAcAucuGuccuuAGT; 3T 800 CuAAGGAcAGAUGUUGGUGT; 3T 801 AD-1 4 19 4 51 % 4 %

AAAGcCcAcuuuAGAGuAuT; 3T 802 AuACUCuAAAGUGGGCUUUT; 3T 803 AD-1 4 195 77 % 5%

AAGcccAcuuuAGAGuAuAT; 3T 804 uAuACUCuAAAGUGGGCUUT; 3T 805 AD-1 4 196 42 % 6%

GAccuuAuuuGGuAAucuGT; 3T 806 cAGAUuACcAAAuAAGGUCT; 3T 807 AD-1 4 197 15% 2 %

GAuuAAuGuAcucAAGAcuT; 3T 808 AGUCUUGAGuAcAUuAAUCT; 3T 809 AD-1 4 198 12 % 2 % cuuuAAGAGGccuAAcuCAT; 3T 810 UGAGUuAGGCCUCUuAAAGT; 3T 811 AD-1 4 199 18 % 2 % uuAAAccAAAcCCuAuuGAT; 3T 812 UcAAuAGGGUUUGGUUuAAT; 3T 813 AD-1 4 200 72 % 9% ucuGuuGGAGAucuAuAAuT; 3T 814 AUuAuAGAUCUCcAAcAGAT; 3T 815 AD-1 4 201 9% 3% cuGAuGuuucuGAGAGAcuT; 3T 816 AGUCUCUcAGAAAcAUcAGT; 3T 817 AD-1 4 202 25% 3%

GcAuAcucuAGucGuucccT; 3T 818 GGGAACGACuAGAGuAUGCT; 3T 819 AD-1 4 203 21 % 1 %

GuuccuuAucGAGAAucuAT; 3T 820 uAGAUUCUCGAuAAGGAACT; 3T 821 AD-1 4 20 4 4 % 2 %

GcAcuuGGAucucucAcAuT; 3T 822 AUGUGAGAGAUCCAAGUGCT; 3T 823 AD-1 4 205 5% 1 %

AAAAAAGGAAcuAGAuGGcT 3T 824 GCcAUCuAGUUCCUUUUUUT 3T 825 AD-1 4 206 79% 6%

SDs single 2nd dose

SEQ SEQ screen

Antisense sequence (5 duplex screen @

Sense sequence (5 -3 )) ID (among 3 ) name 25 nM [ % NO . quadru residual plicat mRNA] es )

AGAGcAGAuuAccucuGcGT; 826 CGcAGAGGuAAUCUGCUCUT; 827 AD-1 4 207 55% 2 %

AGcAGAuuAccucuGcGAGT; 828 CUCGcAGAGGuAAUCUGCUT; 829 AD-1 4 208 100 % 4 %

CCCuGAcAGAGuucAcAAAT; 830 UUUGUGAACUCUGUCAGGGT; 831 AD-1 4 209 34 % 3%

GuuuAccGAAGuGuuGuuuT; 832 AAAcAAcACUUCGGuAAACT; 833 AD-1 4 210 13% 2 % uuAcAGuAcAcAAcAAGGAT; 834 UCCUUGUUGUGUACUGUAAT; 835 AD-1 4 211 9% 1 %

AcuGGAucGuAAGAAGGcAT; 836 UGCCUUCUuACGAUCcAGUT; 837 AD-1 4 212 20 % 3%

GAGcAGAuuAccucuGcGAT; 838 UCGcAGAGGuAAUCUGCUCT; 839 AD-1 4 213 48 % 5%

AAAAGAAGuuAGuGuAcGAT 840 UCGuAcACuAACUUCUUUUT 841 AD-1 4 21 4 28 % 18 %

GAccAuuuAAuuuGGcAGAT 842 UCUGCcAAAUuAAAUGGUCT 843 AD-1 4 215 132 % 0 %

GAGAGGAGuGAuAAuuAAAT 844 UUuAAUuAUCACUCCUCUCT 845 AD-1 4 216 3% 0 % cuGGAGGAuuGGcuGAcAAT 846 UUGUcAGCcAAUCCUCcAGT 847 AD-1 4 217 19% 1 % cucuAGucGuucccAcucAT 848 UGAGUGGGAACGACUAGAGT 849 AD-1 4 218 67 % 8 %

GAuAccAuuAcuAcAGuAGT 850 CuACUGuAGuAAUGGuAUCT 851 AD-1 4 219 76% 4 %

UUCGUCUGCGAAGAAGAAAT 852 UUUCUUCUUCGCAGACGAAT 853 AD-1 4 220 33% 8 %

GAAAAGAAGuuAGuGuAcGT 854 CGuAcACuAACUUCUUUUCT 855 AD-1 4 221 25% 2 % uGAuGuuuAccGAAGuGuuT 856 AAcACUUCGGuAAAcAUcAT 857 AD-1 4 222 7 % 2 % uGuuuGuccAAuucuGGAuT 858 AUCcAGAAUUGGAcAAAcAT 859 AD-1 4 223 19% 2 %

AuGAAGAGuAuAccuGGGAT 860 UCCcAGGuAuACUCUUcAUT 861 AD-1 4 22 4 13% 1 %

GcuAcuCuGAuGAAuGcAuT 862 AUGcAUUcAUcAGAGuAGCT 863 AD-1 4 225 15% 2 %

GcccuuGuAGAAAGAAcAcT 864 GUGUUCUUUCuAcAAGGGCT 865 AD-1 4 226 11 % 0 %

UcAuGuuCcuuAuCGAGAAT 866 UUCUCGAuAAGGAAcAUGAT 867 AD-1 4 227 5% 1 %

GAAuAGGGuuAcAGAGuuGT 868 cAACUCUGuAACCCuAUUCT 869 AD-1 4 228 34 % 3% cAAAcuGGAucGuAAGAAGT 870 CUUCUuACGAUCcAGUUUGT 871 AD-1 4 229 15% 2 % cuuAuuuGGuAAucuGcuGT; 872 cAGcAGAUuACcAAAuAAGT; 873 AD-1 4 230 20 % 1 %

AGcAAuGuGGAAAccuAAcT; 874 GUuAGGUUUCcAcAUUGCUT; 875 AD-1 4 231 18 % 1 %

AcAAuAAAGcAGAcccAuuT; 876 AAUGGGUCUGCUUUAUUGUT; 877 AD-1 4 232 21 % 1 %

AAccAcuuAGuAGuGucCAT; 878 UGGAcACuACuAAGUGGUUT; 879 AD-1 4 233 106% 12 %

AGucAAGAGccAuCuGuAGT; 880 CuAcAGAUGGCUCUUGACUT; 881 AD-1 4 23 4 35% 3% cucccuAGAcuucccuAuuT; 882 AAuAGGGAAGUCuAGGGAGT; 883 AD-1 4 235 48 % 4 %

AuAGcuAAAuuAAAccAAAT; 884 UUUGGUUuAAUUuAGCuAUT; 885 AD-1 4 236 23% 3% uGGcuGGuAuAAuuccAcGT; 886 CGUGGAAUUAUACCAGCCAT; 887 AD-1 4 237 79% 9% uuAuuuGGuAAucuGcuGuT; 888 AcAGcAGAUuACcAAAuAAT; 889 AD-1 4 238 92 % 7 %

AAcuAGAuGGcuuuCucAGT; 890 CUGAGAAAGCcAUCuAGUUT; 891 AD-1 4 239 20 % 2 % ucAuGGcGucGcAGccAAAT; 892 UUUGGCUGCGACGCcAUGAT; 893 AD-1 4 2 4 0 71 % 6%

AcuGGAGGAuuGGcuGAcAT; 894 UGUcAGCcAAUCCUCcAGUT; 895 AD-1 4 2 4 1 14 % 1 % cuAuAAuuGcAcuAucuuuT; 896 AAAGAuAGUGcAAUuAuAGT; 897 AD-1 4 2 4 2 11 % 2 %

AAAGGuCAcCUAAuGAAGAT; 898 UCUUcAUuAGGUGACCUUUT; 899 AD-1 4 2 4 3 11 % 1 %

AuGAAuGcAuAcucuAGucT; 900 GACuAGAGuAUGcAUUcAUT; 901 AD-1 4 2 44 15% 2 %

AAcAuAuuGAAuAAGccuGT; 902 cAGGCUuAUUcAAuAUGUUT; 903 AD-1 4 2 4 5 80 % 7 %

AAGAAGGcAGuuGAccAAcT 904 GUUGGUCAACUGCCUUCUUT 905 AD-1 4 2 4 6 57 % 5%

GAuAcuAAAAGAAcAAuCAT 906 UGAUUGUUCUUUUAGUAUCT 907 AD-1 4 2 4 7 9% 3%

AuAcuGAAAAuCAAuAGuCT 908 GACuAUUGAUUUUcAGuAUT 909 AD-1 4 2 4 8 39% 4 %

AAAAAGGAAcuAGAuGGcuT 910 AGCcAUCuAGUUCCUUUUUT 911 AD-1 4 2 4 9 64 % 2 %

GAAcuAGAuGGcuuuCUCAT 912 UGAGAAAGCcAUCuAGUUCT 913 AD-1 4 250 18 % 2 %

GAAAccuAAcuGAAGAccuT 914 AGGUCUUcAGUuAGGUUUCT 915 AD-1 4 251 56% 6% uAcccAucAAcAcuGGuAAT 916 UuACCAGUGUUGAUGGGuAT 917 AD-1 4 252 48 % 6%

AuuuuGAuAuCuAcccAuuT 918 AAUGGGUAGAUAUCAAAAUT 919 AD-1 4 253 39% 5%

AucccuAuAGuucAcuuuGT 920 cAAAGUGAACuAuAGGGAUT 921 AD-1 4 25 4 44 % 8 %

AuGGGcuAuAAuuGcAcuAT 922 uAGUGcAAUuAuAGCCcAUT 923 AD-1 4 255 108 % 8 %

AGAuuAccucuGcGAGcccT 924 GGGCUCGcAGAGGuAAUCUT 925 AD-1 4 256 108 % 6% uAAuuccAcGuAcccuuCAT 926 UGAAGGGuACGUGGAAUuAT 927 AD-1 4 257 23% 2 %

GucGuucccAcucAGuuuuT 928 AAAACuGAGuGGGAACGACT; 929 AD-1 4 258 21 % 3%

AAAucAAuCCCuGuuGAcuT 930 AGUcAAcAGGGAUUGAUUUT 931 AD-1 4 259 19% 2 %

UCAuAGAGCAAAGAAcAuAT 932 uAUGUUCUUUGCUCuAUGAT 933 AD-1 4 260 10 % 1 % uuAcuAcAGuAGcAcuuGGT; 934 CcAAGUGCuACUGuAGuAAT; 935 AD-1 4 261 76% 3%

AuGuGGAAAccuAAcuGAAT; 936 UUcAGUuAGGUUUCcAcAUT; 937 AD-1 4 262 13% 2 % uGuGGAAAccuAAcuGAAGT; 938 CUUcAGUuAGGUUUCcAcAT; 939 AD-1 4 263 14 % 2 % ucuuccuuAAAuGAAAGGGT; 940 CCCUUUcAUUuAAGGAAGAT; 941 AD-1 4 26 4 65% 3% uGAAGAAccuCuAAGucAAT; 942 UUGACUuAGAGGUUCUUcAT; 943 AD-1 4 265 13% 1 %

AGAGGuCuAAAGuGGAAGAT; 944 UCUUCcACUUuAGACCUCUT; 945 AD-1 4 266 18 % 3%

AuAucuAcccAuuuuucuGT; 946 cAGAAAAAUGGGuAGAuAUT; 947 AD-1 4 267 50 % 9% uAAGccuGAAGuGAAucAGT; 948 CUGAUUcACUUcAGGCUuAT; 949 AD-1 4 268 13% 3%

AGAuGcAGAccAuuuAAuuT; 950 AAUuAAAUGGUCUGcAUCUT; 951 AD-1 4 269 19% 4 %

AGuGuuGuuuGuccAAuucT; 952 GAAUUGGACAAACAACACUT; 953 AD-1 4 270 11 % 2 % cuAuAAuGAAGAGcuuuuuT; 954 AAAAAGCUCUUcAUuAuAGT; 955 AD-1 4 271 11 % 1 %

AGAGGAGuGAuAAuuAAAGT; 956 CUUuAAUuAUcACUCCUCUT; 957 AD-1 4 272 7 % 1 % uuucucuGuuAcAAuAcAuT; 958 AUGuAUUGuAAcAGAGAAAT; 959 AD-1 4 273 14 % 2 %

AAcAuCuAuAAuuGcAAcAT; 960 UGUUGcAAUuAuAGAUGUUT; 961 AD-1 4 27 4 73% 4 %

single SDs

SEQ SEQ dose 2nd

Antisense sequence (5

Sense sequence (5 -3 )) I D ID duplex screen @ screen 3 )

NO . NO . name 25 nM [ % (among residual quadru mRNA] plicat es ) uGcuAGAAGuAcAuAAGAcT; 3T 962 GUCUuAUGuACUUCuAGcAT; 3T 963 AD-1 4 275 10 % 1 %

AAuGuAcucAAGAcuGAucT; 3T 964 GAUcAGUCUUGAGuAcAUUT; 3T 965 AD-1 4 276 89% 2 %

GuAcucAAGAcuGAucuucT; 3T 966 GAAGAUcAGUCUUGAGuACT; 3T 967 AD-1 4 277 7 % 1 % cAcuCuGAuAAAcucAAuGT; 3T 968 cAUUGAGUUuAUcAGAGUGT; 3T 969 AD-1 4 278 12 % 1 %

AAGAGcAGAuuAccucuGcT; 3T 970 GcAGAGGuAAUCUGCUCUUT; 3T 971 AD-1 4 279 104 % 3% ucuGcGAGcccAGAucAAcT; 3T 972 GUUGAUCUGGGCUCGCAGAT; 3T 973 AD-1 4 280 21 % 2 %

AAcuuGAGccuuGuGuAuAT; 3T 974 uAuAcAcAAGGCUcAAGUUT; 3T 975 AD-1 4 281 43% 3%

GAAuAuAuAuAucAGcCGGT 3T 976 CCGGCUGAuAuAuAuAUUCT 3T 977 AD-1 4 282 45% 6% uGucAucccuAuAGuucAcT 3T 978 GUGAACuAuAGGGAUGAcAT 3T 979 AD-1 4 283 35% 5%

GAuCuGGcAAccAuAuuucT 3T 980 GAAAuAUGGUUGCcAGAUCT 3T 981 AD-1 4 28 4 58 % 3% uGGcAAccAuAuuuCuGGAT 3T 982 UCcAGAAAuAUGGUUGCCAT 3T 983 AD-1 4 285 48 % 3%

GAuGuuuAccGAAGuGuuGT 3T 984 cAAcACUUCGGuAAAcAUCT 3T 985 AD-1 4 286 49% 3%

UUCCUuAucGAGAAuCuAAT 3T 986 UuAGAUUCUCGAuAAGGAAT 3T 987 AD-1 4 287 6% 1 %

AGcuuAAuuGcuuucuGGAT 3T 988 UCcAGAAAGcAAUuAAGCUT 3T 989 AD-1 4 288 50 % 2 % uuGcuAuuAuGGGAGAccAT 3T 990 UGGUCUCCcAuAAuAGcAAT 3T 991 AD-1 4 289 48 % 1 %

GucAuGGcGucGcAGccAAT 3T 992 UUGGCUGCGACGCcAUGACT 3T 993 AD-1 4 290 112 % 7 % uAAuuGcAcuAucuuuGcGT 3T 994 CGCAAAGAuAGUGCAAUuAT 3T 995 AD-1 4 291 77 % 2 % cuAucuuuGcGuAuGGccAT 3T 996 UGGCcAuACGcAAAGAuAGT 3T 997 AD-1 4 292 80 % 6% ucccuAuAGuucAcuuuGuT 3T 998 AcAAAGUGAACuAuAGGGAT 3T 999 AD-1 4 293 58 % 2 % ucAAccuuuAAuucAcuuGT 3T 1000 cAAGUGAAUuAAAGGUUGAT 3T 1001 AD-1 4 29 4 77 % 2 %

GGcAAccAuAuuucuGGAAT 3T 1002 UUCcAGAAAuAUGGUUGCCT 3T 1003 AD-1 4 295 62 % 2 %

AuGuAcucAAGAcuGAuCuT 3T 1004 AGAUcAGUCUUGAGuAcAUT 3T 1005 AD-1 4 296 59% 4 %

GcAGAccAuuuAAuuuGGcT 3T 1006 GCcAAAUuAAAUGGUCUGCT 3T 1007 AD-1 4 297 37 % 1 %

UCuGAGAGAcuAcAGAuGuT; 3T 1008 AcAUCUGuAGUCUCUcAGAT; 3T 1009 AD-1 4 298 21 % 1 % uGcucAuAGAGcAAAGAAcT; 3T 1010 GUUCUUUGCUCUAUGAGCAT; 3T 1011 AD-1 4 299 6% 1 %

AcAuAAGAccuuAuuuGGuT; 3T 1012 ACcAAAuAAGGUCUuAUGUT; 3T 1013 AD-1 4 300 17 % 2 % uuuGuGcuGAuucuGAuGGT; 3T 1014 CcAUcAGAAUcAGcAcAAAT; 3T 1015 AD-1 4 301 97 % 6%

CCAucAAcAcuGGuAAGAAT; 3T 1016 UUCUuACCAGUGUUGAUGGT; 3T 1017 AD-1 4 302 13% 1 %

AGAcAAuuCCGGAuGuGGAT; 3T 1018 UCcAcAUCCGGAAUUGUCUT; 3T 1019 AD-1 4 303 13% 3%

GAAcuuGAGccuuGuGuAuT; 3T 1020 AuAcAcAAGGCUcAAGUUCT; 3T 1021 AD-1 4 30 4 38 % 2 % uAAuuuGGcAGAGcGGAAAT; 3T 1022 UUUCCGCUCUGCCAAAUUAT; 3T 1023 AD-1 4 305 14 % 2 % uGGAuGAAGuuAuuAuGGGT; 3T 1024 CCCAuAAuAACUUCAUCCAT; 3T 1025 AD-1 4 306 22 % 4 %

AuCuAcAuGAAcuAcAAGAT; 3T 1026 UCUUGuAGUUcAUGuAGAUT; 3T 1027 AD-1 4 307 26% 6%

GGuAuuuuuGAucuGGcAAT; 3T 1028 UUGCcAGAUcAAAAAuACCT; 3T 1029 AD-1 4 308 62 % 8 %

CuAAuGAAGAGuAuAccuGT; 3T 1030 cAGGuAuACUCUUcAUuAGT; 3T 1031 AD-1 4 309 52 % 5% uuuGAGAAAcuuAcuGAuAT; 3T 1032 uAUcAGuAAGUUUCUcAAAT; 3T 1033 AD-1 4 310 32 % 3%

CGAuAAGAuAGAAGAuCAAT; 3T 1034 UUGAUCUUCuAUCUuAUCGT; 3T 1035 AD-14311 23% 2 % cuGGcAAccAuAuuucuGGT; 3T 1036 CcAGAAAuAUGGUUGCcAGT; 3T 1037 AD-14312 49% 6% uAGAuAccAuuAcuAcAGuT; 3T 1038 ACUGuAGuAAUGGuAUCuAT; 3T 1039 AD-1 4 313 69% 4 %

GuAuuAAAuuGGGuuucAuT 3T 1040 AUGAAACCCAAUUuAAuACT 3T 1041 AD-1 4 31 4 52 % 3%

AAGAccuuAuuuGGuAAucT 3T 1042 GAUuACcAAAuAAGGUCUUT 3T 1043 AD-1 4 315 66% 4 %

GcuGuuGAuAAGAGAGcucT 3T 1044 GAGCUCUCUuAUcAAcAGCT 3T 1045 AD-1 4 316 19% 4 % uAcucAuGuuucucAGAuuT 3T 1046 AAUCUGAGAAACAUGAGUAT 3T 1047 AD-1 4 317 16% 5% cAGAuGGAcGuAAGGcAGcT 3T 1048 GCUGCCUuACGUCcAUCUGT 3T 1049 AD-1 4 318 52 % 11 % uAuCCcAAcAGGuAcGAcAT 3T 1050 UGUCGuACCUGUUGGGAuAT 3T 1051 AD-1 4 319 28 % 11 % cAuuGcuAuuAuGGGAGAcT 3T 1052 GUCUCCcAuAAuAGcAAUGT 3T 1053 AD-1 4 320 52 % 10 %

CCCUCAGuAAAuCCAuGGuT 3T 1054 ACcAUGGAUUuACUGAGGGT 3T 1055 AD-1 4 321 53% 6%

GGucAuuAcuGcccuuGuAT 3T 1056 uAcAAGGGcAGuAAUGACCT 3T 1057 AD-1 4 322 20 % 2 %

AAccAcucAAAAAcAuuuGT 3T 1058 CAAAUGUUUUUGAGUGGUUT 3T 1059 AD-1 4 323 116% 6% uuuGcAAGuuAAuGAAuCuT 3T 1060 AGAUUcAUuAACUUGcAAAT 3T 1061 AD-1 4 32 4 14 % 2 % uuAuuuucAGuAGucAGAAT 3T 1062 UUCUGACuACUGAAAAuAAT 3T 1063 AD-1 4 325 50 % 2 % uuuucucGAuucAAAucuuT 3T 1064 AAGAUUuGAAUCGAGAAAAT; 3T 1065 AD-1 4 326 47 % 3%

GuAcGAAAAGAAGuuAGuGT 3T 1066 cACuAACUUCUUUUCGuACT 3T 1067 AD-1 4 327 18 % 2 % uuuAAAAcGAGAuCUuGcuT 3T 1068 AGcAAGAUCUCGUUUuAAAT 3T 1069 AD-1 4 328 19% 1 %

GAAuuGAuuAAuGuAcucAT; 3T 1070 UGAGuAcAUuAAUcAAUUCT; 3T 1071 AD-1 4 329 94 % 10 %

GAuGGAcGuAAGGcAGcucT; 3T 1072 GAGCUGCCUuACGUCcAUCT; 3T 1073 AD-1 4 330 60 % 4 % cAucuGAcuAAuGGcucuGT; 3T 1074 cAGAGCcAUuAGUcAGAUGT; 3T 1075 AD-1 4 331 54 % 7 %

GuGAuCCuGuAcGAAAAGAT; 3T 1076 UCUUUUCGuAcAGGAUcACT; 3T 1077 AD-1 4 332 22 % 4 %

AGcucuuAuuAAGGAGuAuT; 3T 1078 AuACUCCUuAAuAAGAGCUT; 3T 1079 AD-1 4 333 70 % 10 %

GcucuuAuuAAGGAGuAuAT; 3T 1080 uAuACUCCUuAAuAAGAGCT; 3T 1081 AD-1 4 33 4 18 % 3% ucuuAuuAAGGAGuAuAcGT; 3T 1082 CGuAuACUCCUuAAuAAGAT; 3T 1083 AD-1 4 335 38 % 6% uAuuAAGGAGuAuAcGGAGT; 3T 1084 CUCCGuAuACUCCUuAAuAT; 3T 1085 AD-1 4 336 16% 3%

CuGcAGcccGuGAGAAAAAT; 3T 1086 UUUUUCUcACGGGCUGcAGT; 3T 1087 AD-1 4 337 65% 4 % ucAAGAcuGAucuucuAAGT; 3T 1088 CUuAGAAGAUcAGUCUUGAT; 3T 1089 AD-1 4 338 18 % 0 % cuucuAAGuucAcuGGAAAT; 3T 1090 UUUCcAGUGAACUuAGAAGT; 3T 1091 AD-1 4 339 20 % 4 % uGcAAGuuAAuGAAucuuuT; 3T 1092 AAAGAUUcAUuAACUUGcAT; 3T 1093 AD-1 4 3 4 0 24 % 1 %

AAucuAAGGAuAuAGucAAT; 3T 1094 UUGACuAuAUCCUuAGAUUT; 3T 1095 AD-1 4 3 4 1 27 % 3%

AuCUCuGAAcAcAAGAAcAT; 3T 1096 UGUUCUUGUGUUCAGAGAUT; 3T 1097 AD-1 4 3 4 2 13% 1 %

single SDs

SEQ SEQ dose 2nd

Antisense sequence (5

Sense sequence (5 -3 )) I D ID duplex screen @ screen 3 ) 25 nM [

NO . NO . name % (among residual quadru mRNA] plicat es ) uucuGAAcAGuGGGuAucuT; 3T 1098 AGAuACCcACUGUUCAGAAT; 3T 1099 AD-1 4 3 4 3 19% 1 %

AGuuAuuuAuAcccAucAAT; 3T 1100 UUGAUGGGuAuAAAuAACUT; 3T 1101 AD-1 4 3 44 23% 2 %

AuGcuAAAcuGuucAGAAAT; 3T 1102 UUUCUGAAcAGUUuAGcAUT; 3T 1103 AD-1 4 3 4 5 21 % 4 % cuAcAGAGcAcuuGGuuAcT; 3T 1104 GuAACcAAGUGCUCUGuAGT; 3T 1105 AD-1 4 3 4 6 18 % 2 % uAuAuAucAGccGGGcGcGT; 3T 1106 CGCGCCCGGCUGAUAUAUAT; 3T 1107 AD-1 4 3 4 7 67 % 2 %

AuGuAAAuAcGuAuuuCuAT; 3T 1108 uAGAAAuACGuAUUuAcAUT; 3T 1109 AD-1 4 3 4 8 39% 3% uuuuucucGAuucAAAucuT; 3T 1110 AGAUUuGAAUCGAGAAAAAT 3T 1111 AD-1 4 3 4 9 83% 6%

AAuCUuAAcccuuAGGAcuT 3T 1112 AGUCCuAAGGGUuAAGAUUT 3T 1113 AD-1 4 350 54 % 2 % ccuuAGGAcucuGGuAuuuT 3T 1114 AAAuACcAGAGUCCuAAGGT 3T 1115 AD-1 4 351 57 % 8 %

AAuAAAcuGcccucAGuAAT 3T 1116 UuACUGAGGGcAGUUuAUUT 3T 1117 AD-1 4 352 82 % 3%

GAuCCuGuAcGAAAAGAAGT 3T 1118 CUUCUUUUCGuAcAGGAUCT 3T 1119 AD-1 4 353 2 % 1 %

AAuGuGAuCCUGuAcGAAAT 3T 1120 UUUCGUACAGGAUcAcAUUT 3T 1121 AD-1 4 35 4 18 % 11 %

GuGAAAAcAuuGGccGuucT 3T 1122 GAACGGCcAAUGUUUUcACT 3T 1123 AD-1 4 355 2 % 1 %

CUuGAGGAAAcuCuGAGuAT 3T 1124 uACUcAGAGUUUCCUcAAGT 3T 1125 AD-1 4 356 8 % 2 % cGuuuAAAAcGAGAuCUuGT 3T 1126 cAAGAUCUCGUUUuAAACGT 3T 1127 AD-1 4 357 6% 3% uuAAAAcGAGAuCUuGcuGT 3T 1128 cAGcAAGAUCUCGUUUuAAT 3T 1129 AD-1 4 358 98 % 17 %

AAAGAuGuAuCuGGuCUCCT 3T 1130 GGAGACcAGAuAcAUCUUUT 3T 1131 AD-1 4 359 10 % 1 % cAGAAAAuGuGuCuAcuCAT 3T 1132 UGAGuAGAcAcAUUUUCUGT 3T 1133 AD-1 4 360 6% 4 % cAGGAAuuGAuuAAuGuAcT 3T 1134 GuAcAUuAAUcAAUUCCUGT 3T 1135 AD-1 4 361 30 % 5%

AGucAAcuAAAGcAuAuuuT 3T 1136 AAAuAUGCUUuAGUUGACUT 3T 1137 AD-1 4 362 28 % 2 % uGuGuAAcAAuCuAcAuGAT 3T 1138 UcAUGuAGAUUGUuAcAcAT 3T 1139 AD-1 4 363 60 % 6%

AuAccAuuuGuuccuuGGuT 3T 1140 ACcAAGGAAcAAAUGGuAUT 3T 1141 AD-1 4 36 4 12 % 9%

GcAGAAAuCuAAGGAuAuAT 3T 1142 uAuAUCCUuAGAUUUCUGCT 3T 1143 AD-1 4 365 5% 2 % uGGcuucucAcAGGAAcucT; 3T 1144 GAGUUCCUGUGAGAAGCCAT; 3T 1145 AD-1 4 366 28 % 5%

GAGAuGuGAAuCUCuGAAcT; 3T 1146 GUUcAGAGAUUcAcAUCUCT; 3T 1147 AD-1 4 367 42 % 4 % uGuAAGccAAuGuuGuGAGT; 3T 1148 CUcAcAAcAUUGGCUuAcAT; 3T 1149 AD-1 4 368 93% 12 %

AGccAAuGuuGuGAGGcuuT; 3T 1150 AAGCCUcAcAAcAUUGGCUT; 3T 1151 AD-1 4 369 65% 4 % uuGuGAGGcuucAAGuucAT; 3T 1152 UGAACUUGAAGCCUcAcAAT; 3T 1153 AD-1 4 370 5% 2 %

AGGcAGcucAuGAGAAAcAT; 3T 1154 UGUUUCUcAUGAGCUGCCUT; 3T 1155 AD-1 4 371 54 % 5%

AuAAAuuGAuAGcAcAAAAT; 3T 1156 UUUUGUGCuAUcAAUUuAUT; 3T 1157 AD-1 4 372 4 % 1 %

AcAAAAuCuAGAAcuuAAuT; 3T 1158 AUuAAGUUCuAGAUUUUGUT; 3T 1159 AD-1 4 373 5% 1 %

GAuAuCCcAAcAGGuAcGAT; 3T 1160 UCGuACCUGUUGGGAuAUCT; 3T 1161 AD-1 4 37 4 92 % 6%

AAGuuAuuuAuAcCCAuCAT; 3T 1162 UGAUGGGuAuAAAuAACUUT; 3T 1163 AD-1 4 375 76% 4 % uGuAAAuAcGuAuuucuAGT; 3T 1164 CuAGAAAuACGuAUUuAcAT; 3T 1165 AD-1 4 376 70 % 5% ucuAGuuuucAuAuAAAGuT; 3T 1166 ACUUuAuAUGAAAACUAGAT; 3T 1167 AD-1 4 377 48 % 4 %

AuAAAGuAGuucuuuuAuAT; 3T 1168 uAuAAAAGAACuACUUuAUT; 3T 1169 AD-1 4 378 48 % 3%

CCAuuuGuAGAGcuAcAAAT; 3T 1170 UUUGuAGCUCuAcAAAUGGT; 3T 1171 AD-1 4 379 44 % 5% uAuuuucAGuAGucAGAAuT; 3T 1172 AUUCUGACuACUGAAAAuAT; 3T 1173 AD-1 4 380 35% 16%

AAAuCuAAcccuAGuuGuAT; 3T 1174 uAcAACuAGGGUuAGAUUUT; 3T 1175 AD-1 4 381 44 % 5%

CUUuAGAGuAuAcAuuGcuT 3T 1176 AGcAAUGuAuACUCuAAAGT 3T 1177 AD-1 4 382 28 % 1 %

AucuGAcuAAuGGcucuGuT 3T 1178 AcAGAGCcAUuAGUcAGAUT 3T 1179 AD-1 4 383 55% 11 % cAcAAuGAuuuAAGGAcuGT 3T 1180 cAGUCCUuAAAUcAUUGUGT 3T 1181 AD-1 4 38 4 48 % 9% ucuuuuuCucGAuucAAAuT 3T 1182 AUUuGAAUCGAGAAAAAGAT; 3T 1183 AD-1 4 385 36% 2 % cuuuuucucGAuucAAAucT 3T 1184 GAUUuGAAUCGAGAAAAAGT; 3T 1185 AD-1 4 386 41 % 7 %

AuuuucuGcucAcGAuGAGT 3T 1186 CUcAUCGUGAGcAGAAAAUT 3T 1187 AD-1 4 387 38 % 3% uuucuGcucAcGAuGAGuuT 3T 1188 AACUcAUCGUGAGcAGAAAT 3T 1189 AD-1 4 388 50 % 4 %

AGAGcuAcAAAAcCuAuCCT 3T 1190 GGAuAGGUUUUGuAGCUCUT 3T 1191 AD-1 4 389 98 % 6%

GAGccAAAGGuAcAccAcuT 3T 1192 AGUGGUGuACCUUUGGCUCT 3T 1193 AD-1 4 390 43% 8 %

GccAAAGGuAcAccAcuAcT 3T 1194 GuAGUGGUGuACCUUUGGCT 3T 1195 AD-1 4 391 48 % 4 %

GAAcuGuAcucuucucAGcT 3T 1196 GCUGAGAAGAGuAcAGUUCT 3T 1197 AD-1 4 392 44 % 3%

AGGUAAAUAUcAccAAcAuT 3T 1198 AUGUUGGUGAuAUUuACCUT 3T 1199 AD-1 4 393 37 % 2 %

AGcuAcAAAAccuAuCCUuT 3T 1200 AAGGAuAGGUUUUGuAGCUT 3T 1201 AD-1 4 39 4 114 % 7 % uGuGAAAGcAuuuAAuuCCT 3T 1202 GGAAUuAAAUGCUUUcAcAT 3T 1203 AD-1 4 395 55% 4 %

GcCcAcuuuAGAGuAuAcAT 3T 1204 UGuAuACUCuAAAGUGGGCT 3T 1205 AD-1 4 396 49% 5% uGuGccAcAcuccAAGAccT; 3T 1206 GGUCUUGGAGUGUGGCACAT; 3T 1207 AD-1 4 397 71 % 6%

AAAcuAAAuuGAuCUCGuAT; 3T 1208 uACGAGAUcAAUUuAGUUUT; 3T 1209 AD-1 4 398 81 % 7 % uGAucucGuAGAAuuAucuT; 3T 1210 AGAuAAUUCuACGAGAUcAT; 3T 1211 AD-1 4 399 38 % 4 %

GcGuGcAGucGGuccuccAT; 3T 1212 UGGAGGACCGACUGcACGCT; 3T 1213 AD-I 44 OO 106% 8 %

AAAGuuuAGAGAcAuCuGAT; 3T 1214 UcAGAUGUCUCuAAACUUUT; 3T 1215 AD-1 44 01 47 % 3% cAGAAGGAAuAuGuAcAAAT; 3T 1216 UUUGuAcAuAUUCCUUCUGT; 3T 1217 AD-1 44 02 31 % 1 % cGcccGAGAGuAccAGGGAT; 3T 1218 UCCCUGGuACUCUCGGGCGT; 3T 1219 AD-1 44 03 105% 4 % cGGAGGAGAuAGAAcGuuuT; 3T 1220 AAACGUUCuAUCUCCUCCGT; 3T 1221 AD-I 44 O 4 3% 1 %

AGAuAGAAcGuuuAAAAcGT; 3T 1222 CGUUUuAAACGUUCuAUCUT; 3T 1223 AD-1 44 05 15% 1 %

GGAAcAGGAAcuucAcAAcT; 3T 1224 GUuGuGAAGUUCCuGUUCCT 3T 1225 AD-1 44 06 44 % 5%

GuGAGccAAAGGuAcAccAT; 3T 1226 UGGUGuACCUUUGGCUcACT; 3T 1227 AD-1 44 07 41 % 4 %

AuccucccuAGAcuucccuT; 3T 1228 AGGGAAGUCuAGGGAGGAUT; 3T 1229 AD-1 44 08 104 % 3% cAcAcuccAAGAccuGuGcT; 3T 1230 GcAcAGGUCUUGGAGUGUGT; 3T 1231 AD-1 44 09 67 % 4 %

AcAGAAGGAAuAuGuAcAAT; 3T 1232 UUGuAcAuAUUCCUUCUGUT; 3T 1233 AD-1 44 10 22 % 1 %

SDs single

2nd dose

SEQ SEQ screen

Antisense sequence (5 duplex screen @

Sense sequence (5 -3 ) ID ID (among 3 ) name 25 nM [%

NO. NO. quadru residual plicat mRNA] es) uuAGAGAcAuCuGAcuuuGTsT 1234 cAAAGUcAGAUGUCUCuAATsT 1235 AD-I 44 11 29% 3% AAuuGAuCUcGuAGAAuuATST 1236 uAAUUCuACGAGAUcAAUUTsT 1237 AD-I 44 12 31% 4%

dsRNA targeting the VEGF gene

Four hundred target sequences were identified within exons 1-5 of the VEGF-A121 mRNA sequence, reference transcript is : NM_003376.

1 augaacuuuc ugcugucuug ggugcauugg agccuugccu ugcugcucua ccuccaccau

61 gccaaguggu cccaggcugc acccauggca gaaggaggag ggcagaauca ucacgaagug

121 gugaaguuca uggaugucua ucagcgcagc uacugccauc caaucgagac ccugguggac

181 aucuuccagg aguacccuga ugagaucgag uacaucuuca agccauccug ugugccccug

241 augcgaugcg ggggcugcug caaugacgag ggccuggagu gugugcccac ugaggagucc

301 aacaucacca ugcagauuau gcggaucaaa ccucaccaag gccagcacau aggagagaug

361 agcuuccuac agcacaacaa augugaaugc agaccaaaga aagauagagc aagacaagaa

421 aaaugugaca agccgaggcg guga (SEQ ID NO:1539)

Table 4a includes the identified target sequences. Corresponding siRNAs targeting these sequences were subjected to a bioinformatics screen.

To ensure that the sequences were specific to VEGF sequence and not to sequences from any other genes, the target sequences were checked against the sequences in Genbank using the BLAST search engine provided by NCBI. The use of the BLAST algorithm is described in Altschul et al., J. MoI. Biol. 215:403, 1990; and Altschul and Gish, Meth. Enzymol. 266:460, 1996. siRNAs were also prioritized for their ability to cross react with monkey, rat and human VEGF sequences.

Of these 400 potential target sequences 80 were selected for analysis by experimental screening in order to identify a small number of lead candidates. A total of 114 siRNA molecules were designed for these 80 target sequences 114 (Table 4b).

Table 4a. Target sequences in VEGF- 121

Table 4b: VEGF targeted duplexes Strand: S= sense, AS=Antisense

Example 2. 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 (1 st screen) and 25 nM (2 nd 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-I 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: Effects of Eg5 targeted duplexes on cell viability at 25nM.

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, 0.14 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: IC50 of siRNA: cell proliferation in HeLa cells

Example 3. Eg5 siRNA in vitro screening via mRNA inhibition

Directly before transfection, HeLa S3 (ATCC-Number: CCL-2.2, LCG Promochem GmbH, Wesel, Germany) cells were seeded at 1.5 x 10 4 cells / well on 96-well plates (Greiner Bio-One GmbH, Frickenhausen, Germany) in 75 μl of growth medium (Ham's F 12, 10% fetal calf serum, lOOu penicillin / 100 μg/ml streptomycin, all from Bookroom AG, Berlin, Germany). Transfections were performed in quadruplicates. For each well 0.5 μl Lipofectamine2000 (Invitrogen GmbH, Karlsruhe, 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 % CO 2 in a humidified incubator (Heroes 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 0 C for 30 min. Afterwards, 50 μl of the lists 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 Ib, 2b and 3b).

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 fJVI, 9.9 fJVI 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 Ib, 2b and 3b).

The lead siRNA AD 12115 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 10OnM to 1OfM. 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: Dose res onse mRNA inhibition of E 5/KSP du lexes in HeLa cells

(ND-not determined)

Example 4. Silencing of liver Eg5/KSP in juvenile rats following single-bolus administration of LNPOl 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 using duplex AD-6248..

KSP Duplex Tested

Duplex ID Target Sense Antisense

AD6248 KSP AccGAAGuGuuGuuuGuccTsT (SEQ ID NO 1238 ) GGAcAAAcAAcACUUCGGUTsT (SEQ ID NO 1239)

Methods

Dosing of animals. Male, juvenile Sprague-Dawley rats (19 days old) were administered single doses of lipidoid ("LNPOl") formulated siRNA via tail vein injection. Groups often 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]).

Table 8. Experiment 1

KSP/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.

Example 5. Silencing of rat liver VEGF following intravenous infusion of LNPOl formulated VSP

A "lipidoid" formulation comprising an equimolar mixture of two siRNAs was administered to rats. As used herein, VSP refers to a composition having two siRNAs, one directed to Eg5/KSP and one directed to VEGF. For this experiment the duplex AD3133 directed towards VEGF and AD 12115 directed towards Eg5/KSP were used. Since Eg5/KSP expression is nearly undetectable in the adult rat liver, only VEGF levels were measured following siRNA treatment. siRNA duplexes administered (VSP)

Key: A,G,C,U-ribonucleotides; c,u-2'-O-Me ribonucleotides; s-phosphorothioate. Unmodified versions of each strand and the targets for each siRNA are as follows

unmod sense 5 ' UCGAGAAUCUAAACUAACUTT 3 ' SEQ I D NO : 1534 unmod antisense 3 ' TTAGUCCUUAGAUUUGAUUGA 5 ' SEQ I D NO : 1535

Eg5/KSP target 5 ' UCGAGAAUCUAAACUAACU 3 ' SEQ I D N0 : 1311 unmod sense 5 ' GCACAUAGGAGAGAUGAGCUU 3 ' SEQ I D NO : 1536

VEGF unmod antisense 3 ' GUCGUGUAUCCUCUCUACUCGAA 5 ' SEQ I D NO : 1537 target 5 ' GCACAUAGGAGAGAUGAGCUU 3 ' SEQ I D NO : 1538

Methods

Dosing of animals. Adult, female Sprague-Dawley rats were administered lipidoid ("LNPO 1 ") 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 ND98-4HC1 (MW 1487) (Formula 1, above), Cholesterol (Sigma- Aldrich), and PEG-Ceramide C 16 (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 C 16, 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.

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-XIOO. 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%. For SNALP formulation, the particle size is at least 30 nm, at least 40 nm, at least 50 nm, at least 60 nm, at least 70 nm, at least 80 nm, at least 90 nm, at least 100 nm, at least 110 nm, and at least 120 nm. The preferred range is about at least 50 nm to about at least 110 nm, preferably about at least 60 nm to about at least 100 nm, most preferably about at least 80 nm to about at least 90 nm. In one example, each of the particle size comprises at least about 1 : 1 ratio of Eg5 dsRNA to VEGF dsRNA. 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 Micro BCA 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.

Table 9.

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.

Example 6. Assessment of VSP SNALP in mouse models of human hepatic tumors.

These studies utilized a VSP siRNA cocktail containing dsRNAs targeting KSP/Eg5 and dsRNAs targeting VEGF. As used herein, VSP refers to a composition having two siRNAs, one directed to Eg5/KSP and one directed to VEGF. For this experiment the duplexes AD3133 (directed towards VEGF) and AD 12115 (directed towards Eg5/KSP) were used. The siRNA cocktail was formulated in SNALPs.

The maximum study size utilized 20-25 mice. To test the efficacy of the siRNA SNALP cocktail to treat liver cancer, lxlO λ 6 tumor cells were injected directly into the left lateral lobe of test mice. The incisions were closed by sutures, and the mice allowed to recover for 2-5 hours. The mice were fully recovered within 48-72 hours. The SNALP siRNA treatment was initiated 8-11 days after tumor seeding.

The SNALP formulations utilized were (i) VSP (KSP + VEGF siRNA cocktail (1: 1 molar ratio)); (ii) KSP (KSP + Luc siRNA cocktail); and (iii) VEGF (VEGF + Luc siRNA cocktail). All formulations contained equal amounts (mg) of each active siRNA. All mice received a total siRNA/lipid dose, and each cocktail was formulated into 1:57 cDMA SNALP (1.4% PEG-cDMA; 57.1% DLinDMA; 7.1% DPPC; and 34.3% cholesterol), 6: 1 lipid:drug using original citrate buffer conditions.

Human Hep3B Study A: anti-tumor activity of VSP-SNALP

Human Hepatoma Hep3B tumors were established in scid/beige mice by intrahepatic seeding. Group A (n=6) animals were administered PBS; Group B (n=6) animals were administered VSP SNALP; Group C (n=5) animals were administered KSP/Luc SNALP; and Group D (n=5) animals were administered VEGF/Luc SNALP.

SNALP treatment was initiated eight days after tumor seeding. The SNALP was dosed at 3 mg/kg total siRNA, twice weekly (Monday and Thursday), for a total of six doses (cumulative 18 mg/kg siRNA). The final dose was administered at day 25, and the terminal endpoint was at day 27.

Tumor burden was assayed by (a) body weight; (b) liver weight; (c) visual inspection + photography at day 27; (d) human-specific mRNA analysis; and (e) blood alpha- fetoprotein levels measured at day 27.

Table 10 below illustrates the results of visual scoring of tumor burden measured in the seeded (left lateral) liver lobe. Score: "-" = no visible tumor; "+"= evidence of tumor tissue at injection site; "++" = Discrete tumor nodule protruding from liver lobe; "+++" = large tumor protruding on both sides of liver lobe; "++++" = large tumor, multiple nodules throughout liver lobe.

Table 10.

Liver weights, as percentage of body weight, are shown in FIG. 1.

Body weights are shown in FIGs. 2A-2D.

From this study, the following conclusions were made. (1) VSP SNALP demonstrated potent anti-tumor effects in Hep3B IH model; (2) the anti-tumor activity of the VSP cocktail appeared largely associated with the KSP component; (3) anti-KSP activity was confirmed by single dose histological analysis; and (4) VEGF siRNA showed no measurable effect on inhibition of tumor growth in this model.

Human Hep3B Study B: prolonged survival with VSP treatment

In a second Hep3B study, human hepatoma Hep3B tumors were established by intrahepatic seeding into scid/beige mice. These mice were deficient for lymphocytes and natural killer (NK) cells, which is the minimal scope for immune-mediated anti-tumor effects. Group A (n=6) mice were untreated; Group B (n=6) mice were administered luciferase (luc) 1955 SNALP (Lot No. AP 10-02); and Group C (n=7) mice were administered VSP SNALP (Lot No. APlO-Ol). SNALP was 1:57 cDMA SNALP, and 6: 1 lipid:drug.

SNALP treatment was initiated eight days after tumor seeding. SNALP was dosed at 3 mg/kg siRNA, twice weekly (Mondays and Thursdays), for a total of six doses (cumulative 18 mg/kg siRNA). The final dose was delivered at day 25, and the terminal endpoint of the study was at day 27.

Tumor burden was assayed by (1) body weight; (2) visual inspection + photography at day 27; (3) human-specific mRNA analysis; and (4) blood alpha-fetoprotein measured at day 27.

Body weights were measured at each day of dosing (days 8, 11, 14, 18, 21, and 25) and on the day of sacrifice (FIG. 3).

Table 11.

Score: "-" = no visible tumor; "+"= evidence of tumor tissue at injection site; "++" = Discrete tumor nodule protruding from liver lobe; "+++" = large tumor protruding on both sides of liver lobe; "++++" = large tumor, multiple nodules throughout liver lobe.

The correlation between body weights and tumor burden are shown in FIGs. 4, 5 and 6.

A single dose of VSP SNALP (2 mg/kg) to Hep3B mice also resulted in the formation of mitotic spindles in liver tissue samples examined by histological staining.

Tumor burden was quantified by quantitative RT-PCR (pRT-PCR) (Taqman). Human GAPDH was normalized to mouse GAPDH via species-specific Taqman assays. Tumor score as shown by macroscopic observation in the table above correlated with GADPH levels (FIG. 7A).

Serum ELISA was performed to measure alpha-fetoprotein (AFP) secreted by the tumor. As described below, if levels of AFP go down after treatment, the tumor is not growing. Treatment with VSP lowered AFP levels in some animals compared to treatment with controls (FIG. 7B).

Human HepB3 Study C:

In a third study, human HCC cells (HepB3) were injected directly into the liver of SCID/beige mice, and treatment was initiated 20 days later. Group A animals were administered PBS; Group B animals were administered 4 mg/kg Luc- 1955 SNALP; Group C animals were administered 4 mg/kg SNALP-VSP; Group D animals were administered 2 mg/kg SNALP-VSP; and Group E animals were administered 1 mg/kg SNALP-VSP. Treatment was with a single intravenous (iv) dose, and mice were sacrificed 24 hr. later.

Tumor burden and target silencing was assayed by qRT-PCR (Taqman). Tumor score was also measured visually as described above, and the results are shown in the following table. hGAPDH levels, as shown in FIG. 8, correlates with macroscopic tumor score as shown in the table below. Table 12.

Score: "+"= variable tumor take/ some small tumors; "++" = Discrete tumor nodule protruding from liver lobe; "+++" = large tumor protruding on both sides of liver lobe

Human (tumor-derived) KSP silencing was assayed by Taqman analysis and the results are shown in FIG. 10. hKSP expression was normalized to hGAPDH. About 80% tumor KSP silencing was observed at 4 mg/kg SNALP-VSP, and efficacy was evident at 1 mg/kg. The clear bars in FIG. 9 represent the results from small (low GAPDH) tumors.

Human (tumor-derived) VEGF silencing was assayed by Taqman analysis and the results are shown in FIG. 10. hVEGF expression was normalized to hGAPDH. About 60% tumor VEGF silencing was observed at 4 mg/kg SNALP-VSP, and efficacy was evident at 1 mg/kg. The clear bars in FIG. 10 represent the results from small (low GAPDH) tumors.

Mouse (liver-derived) VEGF silencing was assayed by Taqman analysis and the results are shown in FIG. 1 IA. mVEGF expression was normalized to hGAPDH. About 50% liver VEGF silencing was observed at 4 mg/kg SNALP-VSP, and efficacy was evident at 1 mg/kg.

Human HepB3 Study D: contribution of each dsRNA to tumor growth

In a fourth study, human HCC cells (HepB3) were injected directly into the liver of SCID/beige mice, and treatment was initiated 8 days later. Treatment was with intravenous (iv) bolus injections, twice weekly, for a total of six does. The final dose was administered at day 25, and the terminal endpoint was at day 27.

Tumor burden was assayed by gross histology, human-specific mRNA analysis (hGAPDH qPCR), and blood alpha-fetoprotein levels (serum AFP via ELISA).

In Study 1, Group A was treated with PBS, Group B was treated with SNALP- KSP+Luc (3 mg/kg), Group C was treated with SNALP -VEGF+Luc (3 mg/kg), and Group D was treated with ALN-VSP02 (3 mg/kg).

In Study 2, Group A was treated with PBS; Group B was treated with SNALP- KSP+Luc (1 mg/kg), Group C was treated with ALN-VSP02 (1 mg/kg).

Both GAPDH mRNA levels and serum AFP levels were shown to decrease after treatment with ALN-VSP02 (FIG. 1 IB).

Histology Studies:

Human hepatoma Hep3B tumors were established by intrahepatic seeding in mice. SNALP treatment was initiated 20 days after tumor seeding. Tumor-bearing mice (three per group) were treated with a single intravenous (IV) dose of (i) VSP SNALP or (ii) control (Luc) SNALP at 2 mg/kg total siRNA.

Liver/tumor samples were collected for conventional H&E histology 24 hours after single SNALP administration.

Large macroscopic tumor nodules (5-10 mm) were evident at necroscopy.

Effect of ALN-VSP in Hep3B mice:

ALN-VSP (a cocktail of KSP dsRNA and VEGF dsRNA) treatment reduced tumor burden and expression of tumor-derived KSP and VEGF. GAPDH mRNA levels, a measure of tumor burden, were also observed to decline following administration of ALN-VSP dsRNA (see FIGs. 12A-12C). A decrease in tumor burden by visual macroscopic observation was also evident following administration of ALN-VSP.

A single IV bolus injection of ALN-VSP also resulted in mitotic spindle formation that was clearly detected in liver tissue samples from Hep3B mice. This observation indicated cell cycle arrest.

Example 7. Survival of SNALP-VSP animals versus SNALP-Luc treated animals

To test the effect of siRNA SNALP on survival rates of cancer subjects, tumors were established by intrahepatic seeding in mice and the mice were treated with SNALP-siRNA. These studies utilized a VSP siRNA cocktail containing dsRNAs targeting KSP/Eg5 and VEGF. Control was dsRNA targeting Luc. The siRNA cocktail was formulated in SNALPs.

Tumor cells (Human Hepatoma Hep3B, lxlO λ 6) were injected directly into the left lateral lobe of scid/beige mice. These mice were deficient for lymphocytes and natural killer (NK) cells, which is the minimal scope for immune-mediated anti-tumor effects. The incisions were closed by sutures, and the mice allowed to recover for 2-5 hours. The mice were fully recovered within 48-72 hours.

All mice received a total siRNA/lipid intravenous (iv) dose, and each cocktail was formulated into 1:57 cDMA SNALP (1.4% PEG-cDMA; 57.1% DLinDMA; 7.1% DPPC; and 34.3% cholesterol), 6: 1 lipid:drug using original citrate buffer conditions. siRNA-SNALP treatment was initiated on the day indicated below (18 or 26 days) after tumor seeding. siRNA-SNALP were administered twice a week for three weeks after at a dose of 4 mg/kg. Survival was monitored and animals were euthanized based on humane surrogate endpoints (e.g., animal body weight, abdominal distension/discoloration, and overall health).

The survival data for treatment initiated 18 days after tumor seeing is summarized in Table 13, Table 14, and FIG. 13 A.

Table 13. Kaplan-Meier (survival) data (% Surviving)

Table 14. Survival in days, for each animal.

FIG. 13 A shows the mean survival of SNALP-VSP animals and SNALP-Luc treated animals versus days after tumor seeding. The mean survival of SNALP-VSP animals was extended by approximately 15 days versus SNALP-Luc treated animals.

Table 15. Serum alpha fetoprotein (AFP) concentration, for each animal, at a time pre-treatment and at end of treatment (concentration in μg/ml)

Tumor burden was monitored using serum AFP levels during the course of the experiment. Alpha-fetoprotein (AFP) is a major plasma protein produced by the yolk sac and the liver during fetal life. The protein is thought to be the fetal counterpart of serum albumin, and human AFP and albumin gene are present in tandem in the same transcriptional

orientation on chromosome 4. AFP is found in monomeric as well as dimeric and trimeric forms, and binds copper, nickel, fatty acids and bilirubin. AFP levels decrease gradually after birth, reaching adult levels by 8-12 months. Normal adult AFP levels are low, but detectable. AFP has no known function in normal adults and AFP expression in adults is often associated with a subset of tumors such as hepatoma and teratoma. AFP is a tumor marker used to monitor testicular cancer, ovarian cancer, and malignant teratoma. Principle tumors that secrete AFP include endodermal sinus tumor (yolk sac carcinoma), neuroblastoma, hepatoblastoma, and heptocellular carcinoma. In patients with AFP-secreting tumors, serum levels of AFP often correlate with tumor size. Serum levels are useful in assessing response to treatment. Typically, if levels of AFP go down after treatment, the tumor is not growing. A temporary increase in AFP immediately following chemotherapy may indicate not that the tumor is growing but rather that it is shrinking (and releasing AFP as the tumor cells die). Resection is usually associated with a fall in serum levels. As shown in Figure 14, tumor burden in SNALP-VSP treated animals was significantly reduced.

The experiment was repeated with SNALP-siRNA treatment at 26, 29, 32 35, 39, and 42 days after implantation. The data is shown in FIG. 13B. The mean survival of SNALP- VSP animals was extended by approximately 15 days versus SNALP -Luc treated animals by approximately 19 days, or 38%.

Example 8. Induction of Mono-asters in Established Tumors

Inhibition of KSP in dividing cells leads to the formation of mono asters that are readily observable in histological sections. To determine whether mono aster formation occurred in SNALP-VSP treated tumors, tumor bearing animals (three weeks after Hep3B cell implantation) were administered 2 mg/kg SNALP-VSP via tail vein injection. Control animals received 2 mg/kg SNALP -Luc. Twenty four hours later, animals were sacrificed, and tumor bearing liver lobes were processed for histological analysis. Representative images of H&E stained tissue sections are shown in Figure 15. Extensive mono aster formation was evident in ALN VSP02 treated (A), but not SNALP-Luc treated (B), tumors. In the latter, normal mitotic figures were evident. The generation of mono asters is a characteristic feature of KSP inhibition and provides further evidence that SNALP-VSP has significant activity in established liver tumors.

Example 9. Manufacturing Process and Product specification of ALN-VSP02 (SNALP-VSP)

ALN-VSP02 product contains 2 mg/mL of drug substance ALN-VSPDSOl formulated in a sterile lipid particle formulation (referred to as SNALP) for IV administration

via infusion. Drug substance ALN-VSPDSOl consists of two siRNAs (ALN- 12115 targeting KSP and ALN-3133 targeting VEGF) in an equimolar ratio. The drug product is packaged in 10 mL glass vials with a fill volume of 5 mL.

The following terminology is used herein:

*Altemate names = AD-12115, AD12115; ** Alternate names = AD-3133, AD3133

9.1 Preparation of drug substance ALN-VSPDSOl

The two siRNA components of drug substance ALN-VSPDSOl, ALN- 12115 and ALN-3133, are chemically synthesized using commercially available synthesizers and raw materials. The manufacturing process consists of synthesizing the two single strand oligonucleotides of each duplex (A 19562 sense and A 19563 antisense of ALN 12115 and A 3981 sense and A 3982 antisense of ALN 3133) by conventional solid phase oligonucleotide synthesis using phosphoramidite chemistry and 5' O dimethoxytriphenylmethyl (DMT) protecting group with the 2' hydroxyl protected with tert butyldimethylsilyl (TBDMS) or the 2' hydroxyl replaced with a 2' methoxy group (2' OMe). Assembly of an oligonucleotide chain by the phosphoramidite method on a solid support such as controlled pore glass or polystyrene. The cycle consists of 5' deprotection, coupling, oxidation, and capping. Each coupling reaction is carried out by activation of the appropriately protected ribo , 2' OMe , or deoxyribonucleoside amidite using 5 (ethylthio) IH tetrazole reagent followed by the coupling of the free 5' hydroxyl group of a support immobilized protected nucleoside or oligonucleotide. After the appropriate number of cycles, the final 5' protecting group is removed by acid treatment. The crude oligonucleotide is cleaved from the solid support by aqueous methylamine treatment with concomitant removal of the cyanoethyl protecting group as well as nucleobase protecting groups. The 2' O TBDMS group is then cleaved using a hydrogen fluoride containing reagent to yield the crude oligoribonucleotide, which is purified using strong anion exchange high performance liquid chromatography (HPLC) followed by desalting using ultrafiltration. The purified single strands are analyzed to confirm the correct molecular weight, the molecular sequence, impurity profile and oligonucleotide content, prior to annealing into the duplexes. The annealed duplex intermediates ALN 12115 and ALN 3133 are either lyophilized and stored at 20 0 C or mixed in 1: 1 molar ratio and the solution is lyophilized to yield drug substance ALN VSPDSOl. If the duplex intermediates were stored

as dry powder, they are redissolved in water before mixing. The equimolar ratio is achieved by monitoring the mixing process by an HPLC method.

The manufacturing process flow diagram is shown in Figure 16.

Example specifications are shown in Table 16a.

The results of up to 12 month stability testing for ALN-VSPDSOl drug substance are shown in Tables 16c. The assay methods were chosen to assess physical property (appearance, pH, moisture), purity (by SEC and denaturing anion exchange chromatography) and potency (by denaturing anion exchange chromatography [AX-HPLC]).

Table 16a. Example specifications for ALN-VSPDSOl

Table 16b: Stability of drug substance

9.2 Preparation of drug product ALN-VSP02 (SNALP-VSP) ALN VSP02, is a sterile formulation of the two siRNAs (in a 1: 1 molar ratio) with lipid excipients in isotonic buffer. The lipid excipients associate with the two siRNAs, protect them from degradation in the circulatory system, and aid in their delivery to the target tissue. The specific lipid excipients and the quantitative proportion of each (shown in Table 17) have been selected through an iterative series of experiments comparing the physicochemical properties, stability, pharmacodynamics, pharmacokinetics, toxicity and product manufacturability of numerous different formulations. The excipient DLinDMA is a titratable aminolipid that is positively charged at low pH, such as that found in the endosome of mammalian cells, but relatively uncharged at the more neutral pH of whole blood. This feature facilitates the efficient encapsulation of the negatively charged siRNAs at low pH, preventing formation of empty particles, yet allows for adjustment (reduction) of the particle charge by replacing the formulation buffer with a more neutral storage buffer prior to use. Cholesterol and the neutral lipid DPPC are incorporated in order to provide physicochemical stability to the particles. The polyethyleneglycol lipid conjugate PEG2000 C DMA aids drug product stability, and provides optimum circulation time for the proposed use. ALN VSP02 lipid particles have a mean diameter of approximately 80-90 nm with low polydispersity values. A representative cryo transmission electron microscope (cryo TEM) image is shown in Figure 17. At neutral pH, the particles are essentially uncharged, with

Zeta Potential values of less than 6 mV. There is no evidence of empty (non loaded) particles based on the manufacturing process.

Table 17: Quantitative Composition of ALN-VSP02

* The 1 : 1 molar ratio of the two siRNAs in the drug product is maintained throughout the size distribution of the drug product particles.

Solutions of lipid (in ethanol) and ALN VSPDSOl drug substance (in aqueous buffer) are mixed and diluted to form a colloidal dispersion of siRNA lipid particles with an average particle size of approximately 80-90 nm. This dispersion is then filtered through 0.45/0.2 μm filters, concentrated, and diafiltered by Tangential Flow Filtration. After in process testing and concentration adjustment to 2.0 mg/mL, the product is sterile filtered, aseptically filled into glass vials, stoppered, capped and placed at 5 ± 3 0 C. The ethanol and all aqueous buffer components are USP grade; all water used is USP Sterile Water For Injection grade. Representative ALN-VSP02 process is shown in flow diagram in FIG. 18.

Table 18a: Example ALN-VSP02 specifications

9.4 Container/Closure System

The ALN VSP02 drug product is packaged in 10 mL glass vials with a fill volume of 5 mL. The container closure system is comprised of a USP/EP Type I borosilicate glass vial, a teflon faced butyl rubber stopper and an aluminum flip off cap. The drug product will be stored at 5 ± 3°C.

9.5 Stability of drug product ALN-VSP02

Stability data (25°C/60%RH) are given in Table 18b and 18c. Table 18b: Example ALN-VSP02 stability at storage conditions

Table 18c: Example ALN-VSP02 stability at 25°C/ambient humidity

Example 10. In Vitro Efficacy of ALN-VSP02 in Human Cancer Cell Lines

The efficacy of ALN-VSP02 treatment in human cancer cell lines was determined via measurement of KSP mRNA, VEGF mRNA, and cell viability after treatment. IC50 (nM) values determined for KSP and VEGF in each cell line.

Table 19: cell lines

Cell line tested ATCC cat number HELA ATCC Cat N: CCL-2

KB ATCC Cat N: CCL- 17

HEP3B ATCC Cat N: HB-8064

SKOV-3 ATCC Cat N: HTB-77

HCT-116 ATCC Cat N: CCL-247

HT-29 ATCC Cat N: HTB-38

PC-3 ATCC Cat N: CRL-1435

A549 ATCC CatN: CCL-185

MDA-MB-231 ATCC Cat N: HTB-26

Cells were plated in 96 well plates in complete media at day 1 to reach a density of 70% on day 2. On day 2 media was replaced with Opti-MEM reduced serum media (Invitrogen Cat N: 11058-021) and cells were transfected with either ALN-VSP02 or control SNALP-Luc with concentration range starting at 1.8 μM down to 10 pM. After 6 hours the media was changed to complete media. Three replicate plates for each cell line for each experiment was done.

Cells were harvested 24 hours after transfection. KSP levels were measured using bDNA; VEGF mRNA levels were measured using human TaqMan assay.

Viability was measured using Cell Titer Blue reagent (Promega Cat N: G8080) at 48 and/or 72h following manufacturer's recommendations.

As shown in Table 20, nM concentrations of VSP02 are effective in reducing expression of both KSP and VEGF in multiple human cell lines. Viability of treated cells was not

Table 20: Results

Example 11. Anti-tumor efficacy of VSP SNALP vs. Sorafenib in established Hep3B intrahepatic tumors

The anti-tumor effects of multi-dosing VSP SNALP verses Sorafenib in scid/beige mice bearing established Hep3B intrahepatic tumors was studied. Sorafenib is a small molecule inhibitor of protein kinases approved for treatment of hepatic cellular carcinoma (HCC).

Tumors were established by intrahepatic seeding in scid/beige mice as described herein. Treatment was initiated 11 days post-seeding. Mice were treated with Sorafenib and a control siRNA-SNALP, Sorafenib and VSP siRNA-SNALP, or VSP siRNA-SNALP only. Control mice were treated with buffers only (DMSO for Sorafenib and PBS for siRNA- SNALP). Sorafenib was administered intraparenterally from Mon to Fri for three weeks, at 15 mg/kg according to body weight for a total of 15 injections. Sorafenib was administered a minimum of 1 hour after SNALP injections. The siRNA-SNALPS were administered intravenously via the lateral tail vein according at 3 mg/kg based on the most recently recorded body weight (10 ml/kg) for 3 weeks (total of 6 doses) on days 1, 4, 7, 10, 14, and 17.

Mice were euthanized based on an assessment of tumor burden including progressive weight loss and clinical signs including condition, abdominal distension/discoloration and mobility.

The percent survival data are shown in FIG. 21. Co-administration of VSP siRNA- SNALP with Sorafenib increased survival proportion compared to administration of Sorafenib or VSP siRNA-SNALP alone. VSP siRNA-SNALP increased survival proportion compared to Sorafenib.

Example 12. In vitro efficacy of VSP using variants of AD-12115 and AD-3133

Two sets of duplexes targeted to Eg5/KSP and VEGF were designed and synthesized. Each set included duplexes tiling 10 nucleotides in each direction of the target sites for either AD-12115 and AD-3133.

Sequences of the target, sense strand, and antisense strand for each duplex are shown in the Table below.

Each duplex is assayed for inhibition of expression using the assays described herein. The duplexes are administered alone and/or in combination, e.g., an Eg5/KSP dsRNA in combination with a VEGF dsRNA. In some embodiments, the dsRNA are administered in a SNALP formulation as described herein.

Table 21 : Sequences of dsRNA targeted to VEGF and Eg5/KSP (tiling)

Example 13. VEGF targeted dsRNA with a single blunt end

A set duplexes targeted to VEGF were designed and synthesized. The set included duplexes tiling 10 nucleotides in each direction of the target sites for AD-3133. Each duplex includes a 2 base overhang at the end corresponding to the 3' end of the antisense strand and no overhang, e.g., a blunt end, at the end corresponding to the 5' end of the antisense strand.

The sequences of each strand of these duplexes are shown in the following table.

Each duplex is assayed for inhibition of expression using the assays described herein. The VEGF duplexes are administered alone and/or in combination with an Eg5/KSP dsRNA (e.g., AD-12115). In some embodiments, the dsRNA are administered in a SNALP formulation as described herein.

Table 22: Target sequences of blunt ended dsRNA targeted to VEGF

Table 23: Strand sequences of blunt ended dsRNA targeted to VEGF

Example 14. Inhibition of Eg5/KSP and VEGF expression in humans

A human subject is treated with a pharmaceutical composition, e.g., ALNVSP02, having both a SNALP formulated dsRNA targeted to a Eg5/KSP gene and a SNALP formulated dsRNA targeted to a VEGF gene to inhibit expression of the Eg5/KSP and VEGF genes.

A subject in need of treatment is selected or identified. The subject can be in need of cancer treatment, e.g., liver cancer.

At time zero, a suitable first dose of the composition is subcutaneously administered to the subject. The composition is formulated as described herein. After a period of time, the subject's condition is evaluated, e.g., by measurement of tumor growth, measuring serum AFP levels, and the like. This measurement can be accompanied by a measurement of Eg5/KSP and/or VEGF expression in said subject, and/or the products of the successful

siRNA-targeting of Eg5/KSP and/or VEGF mRNA. Other relevant criteria can also be measured. The number and strength of doses are adjusted according to the subject's needs.

After treatment, the subject's condition is compared to the condition existing prior to the treatment, or relative to the condition of a similarly afflicted but untreated subject.

Those skilled in the art are familiar with methods and compositions in addition to those specifically set out in the present disclosure which will allow them to practice this invention to the full scope of the claims hereinafter appended.