ENHANCED ANTISENSE OLIGONUCLEOTIDES

Described herein are gap-widened antisense oligonucleotides having improved therapeutic index as compared to 5-10-5 MOE gapmer antisense oligonucleotides of the same sequence. Also described are methods of reducing a target RNA in an animal using the gap-widened antisense oligonucleotides of the present invention. Further, are methods for selecting a gap-widened antisense oligonucleotides.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. application Ser. No. 11/231,243, filed Sep. 19, 2005, allowed Oct. 19, 2010, which claims priority under 35 USC 119(e) to U.S. Application No. 60/611,100, filed on Sep. 17, 2004 and to U.S. Application No. 60/663,442, filed on Mar. 18, 2005, each of which is herein incorporated by reference in its entirety. The instant application is also related to U.S. Application 60/718,685, filed Sep. 19, 2005, and U.S. Application 60/718,684, filed Sep. 19, 2005, each of which is herein incorporated by reference in its entirety.

SEQUENCE LISTING

The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled CORE0051USC1SEQ.txt, created on Jan. 17, 2011 which is 92 Kb in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.

FIELD OF THE INVENTION

The present invention provides chimeric antisense compounds having enhanced in vivo potency and thus an improved therapeutic index. The compounds described herein have widened deoxy gaps and enhanced in vivo potency which is unexpected based on their in vitro activity.

BACKGROUND OF THE INVENTION

Antisense oligonucleotides are accepted therapeutic modalities and many thousands of patients have been treated with antisense compounds. The original “first generation” antisense compounds employed in the first antisense clinical trials were oligodeoxynucleotides having 2′-deoxy ribonucleotides and phosphorothioate intemucleoside linkages. Subsequently, chimeric “second generation” antisense oligonucleotides exhibited a marked improvement in potency over first generation antisense oligonucleotides. Second generation antisense oligonucleotides are chimeric oligonucleotides typically having a 2′-deoxy “gap” region flanked by “wings” having nucleotides with 2′-modified ribonucleotides, referred to as “gapmers.” The most widely used of the “second generation” antisense motifs is often referred to as a “MOE gapmer” in which the 2′-modified ribonucleotide is a 2′-O-methoxyethyl (2′-MOE or simply MOE) modification, and each of the internucleotide linkages is a phosphorothioate. Predominantly, second generation oligonucleotides have a length of 20 nucleotides of which the 5 nucleotides at each terminus are 2′-MOE nucleotides and the center ten nucleotides are 2′-deoxyribonucleotides. These second generation oligonucleotides are referred to as “5-10-5 MOE gapmers” have a 5-10-5 wing-gap-wing motif. Chimeric antisense compounds with other arrangements of modifications have also been made. “Hemimers,” are chimeric compounds in which there is a single 2′-modified “wing” adjacent to (on either the 5′, or the 3′ side of) a 2′-deoxy gap have been described (Geary et al., 2001, J. Pharm. Exp. Therap., 296, 898-904).

SUMMARY OF THE INVENTION

The present invention is directed to “gap-widened” antisense oligonucleotides having a gap region of greater than 11 2′deoxyribonucleotides flanked by two “wing” regions having from one to eight nucleotides which do not support RNase H activity. The gap-widened antisense oligonucleotide of the present invention have been shown to have an improved therapeutic index as compared to a corresponding antisense oligonucleotide having a 5-10-5 MOE gamer antisense oligonucletide with the same sequence. The gap-widened antisense oligonucleotides of the present invention exhibit increased in vivo potency or improved tissue exposure as compared with the corresponding 5-10-5 MOE gapmer antisense oligonucleotide with the same sequence. Most interestingly, there is a lack of correlation between the in vitro potency and the in vivo potency of the gap-widened antisense oligonucleotides described herein. The gap-widened antisense oligonucleotides of the present invention are 18 to 24 nucleotides in length. In particular, the gap-widened antisense oligonucleotides of the present invention have wing regions having 2′-O-(2-methoxyethyl)ribonucleotides.

In an additional embodiment of the present invention is a method of reducing expression of a target RNA in an animal in need of reducing expression of said target RNA, comprising administering to said animal a gap-widened antisense oligonucleotide 18 to 24 nucleotides in length comprising: a gap region having greater than 11 contiguous 2′-deoxyribonucleotides; and a first wing region and a second wing region flanking the gap region, wherein each of said first and second wing regions independently have 1 to 8 2′-O-(2-methoxyethyl)ribonucleotides, having an improved therapeutic index as compared to a corresponding 5-10-5 MOE gapmer antisense oligonucleotide having a gap region of 10 contiguous 2′-deoxyribonucleotides and a first wing region and a second wing region flanking the gap region of 5 2′-O-(2-methoxyethyl)ribonucleotides. The improvement in therapeutic index is characterized by equal or increased potency coupled with a reduction in tissue concentration, or increased potency coupled with equal tissue exposures as compared to a corresponding 5-10-5 MOE gapmer antisense oligonucleotide of the same sequence. In addition, the improvement in therapeutic index may be characterized by an increased liver to kidney concentration ratio as compared to a corresponding 5-10-5 MOE gapmer antisense oligonucleotide of the same sequence. In particular, the method of the present invention is useful in reducing the expression of RNA targets expressed in the kidney, liver, or adipose tissues. The method of the present invention is also useful in reducing the expression of target RNA associated with a metabolic or cardiovascular disease or condition. The method of the present invention is useful wherein the metabolic disease or condition is selected from diabetes, hepatic steatosis, fatty liver disease, non-alcoholic steatohepatitis, metabolic syndrome, obesity, or the like. In addition, the method of the present invention is useful wherein the cardiovascular disease or condition is selected from hypercholesterolemia, atherosclerosis, hyperlipidemia, familial hypercholesterolemia, or the like.

An additional method of the present invention is a method of selecting a gap-widened antisense oligonucleotide with an improved therapeutic index, the method comprising:

screening in vitro a plurality of antisense oligonucleotides targeting a human RNA and having a single wing-gap-wing motif;

identifying a parent antisense oligonucleotide from the plurality of antisense oligonucleotides having a potent in vitro activity;

synthesizing a plurality of gap-widened antisense oligonucleotides having the same sequence as the parent antisense oligonucleotide, wherein said gap-widened antisense oligonucleotide is 18 to 24 nucleotides in length comprising a gap region having greater than 11 contiguous 2′-deoxyribonucleotides; and a first wing region and a second wing region flanking the gap region, wherein each of said first and second wing regions independently has 1 to 8 2′-O-(2-methoxyethyl)ribonucleotides;

testing said plurality of gap-widened antisense oligonucleotides in a plurality of animals;

obtaining potency and tissue concentration data from said testing step; and

determining an optimized gap-widened oligonucleotide wing-gap-wing motif with an improved therapeutic index, improved potency, reduced tissue exposure, or reduced toxicity, or a combination thereof as compared to the parent antisense oligonucleotide.

In one embodiment, the method of selecting a gap-widened antisense oligonucleotide further comprises the step of designing a rodent sequence analogous or a non-human primate sequence to said parent antisense oligonucleotide. In one embodiment, the step of determining the optimized gap-widened antisense oligonucleotide wing-gap-wing motif with an improved therapeutic index includes identifying a gap-widened antisense oligonucleotide which has equal or increased potency as compared to the parent antisense oligonucleotide.

In the step of screening, each of said antisense oligonucleotides has the same wing-gap-wing motif selected from 2-16-2, 3-14-3, 4-12-4, or 5-10-5. In a further embodiment, the wing portions of the gap-widened antisense oligonucleotides are 2′-O-(2-methoxyethyl)ribonucleotides. In particular, the step of screening is performed in primary hepatocytes, HepG2, bEND, or HeLa cells. In the step of identifying, the potent in vitro activity is greater than 50% reduction in the target mRNA expression as compared to a saline control. In alternate embodiments, in the step of identifying, the potent in vitro activity is greater than 30%, greater than 40%, greater than 50%, greater than 60%, greater than 70%, greater than 80%, or greater than 90%

In the step of synthesizing, the gap-widened antisense oligonucleotides each have different wing-gap-wing motifs. In particular, the gap-widened antisense oligonucleotides have gaps of 12, 13, 14, 15, 16, 17, or 18 2′-deoxyribonucleotides in length. In the step of testing, the animals are selected from rodents such as mice and rats, and non-human primates, such as cynomolgous monkeys.

In the step of obtaining, the tissue concentration data are concentrations of full-length gap-widened antisense oligonucleotides particularly measured in the liver, kidney, or adipose tissue. In one embodiment, each optimized gap-widened antisense oligonucleotide is selected because of equal or improved potency data. In another embodiment, each optimized gap-widened antisense oligonucleotide is selected because of reduced tissue exposure. In another embodiment, each optimized gap-widened antisense oligonucleotide is selected because of reduced toxicity. In another embodiment, each optimized gap-widened antisense oligonucleotide is selected because of improved therapeutic index. In another embodiment, each optimized gap-widened antisense oligonucleotide is selected because of reduced tissue exposure, reduced toxicity, improved potency, or a combination thereof.

The gap-widened antisense oligonucleotides described herein may have various wing-gap-wing motifs selected from: 1-16-1, 2-15-1, 1-15-2, 1-14-3, 3-14-1, 2-14-2, 1-13-4, 4-13-1, 2-13-3, 3-13-2, 1-12-5, 5-12-1, 2-12-4, 4-12-2, 3-12-3, 1-11-6, 6-11-1, 2-11-5, 5-11-2, 3-11-4, 4-11-3, 1-17-1, 2-16-1, 1-16-2, 1-15-3, 3-15-1, 2-15-2, 1-14-4, 4-14-1, 2-14-3, 3-14-2, 1-13-5, 5-13-1, 2-13-4, 4-13-2, 3-13-3, 1-12-6, 6-12-1, 2-12-5, 5-12-2, 3-12-4, 4-12-3, 1-11-7, 7-11-1, 2-11-6, 6-11-2, 3-11-5, 5-11-3, 4-11-4, 1-18-1, 1-17-2, 2-17-1, 1-16-3, 1-16-3, 2-16-2, 1-15-4, 4-15-1, 2-15-3, 3-15-2, 1-14-5, 5-14-1, 2-14-4, 4-14-2, 3-14-3, 1-13-6, 6-13-1, 2-13-5, 5-13-2, 3-13-4, 4-13-3, 1-12-7, 7-12-1, 2-12-6, 6-12-2, 3-12-5, 5-12-3, 4-12-4, 1-11-8, 8-11-1, 2-11-7, 7-11-2, 3-11-6, 6-11-3, 4-11-5, 5-11-4, 1-18-1, 1-17-2, 2-17-1, 1-16-3, 3-16-1, 2-16-2, 1-15-4, 4-15-1, 2-15-3, 3-15-2, 1-14-5, 2-14-4, 4-14-2, 3-14-3, 1-13-6, 6-13-1, 2-13-5, 5-13-2, 3-13-4, 4-13-3, 1-12-7, 7-12-1, 2-12-6, 6-12-2, 3-12-5, 5-12-3, 4-12-4, 1-11-8, 8-11-1, 2-11-7, 7-11-2, 3-11-6, 6-11-3, 4-11-5, 5-11-4, 1-19-1, 1-18-2, 2-18-1, 1-17-3, 3-17-1, 2-17-2, 1-16-4, 4-16-1, 2-16-3, 3-16-2, 1-15-5, 2-15-4, 4-15-2, 3-15-3, 1-14-6, 6-14-1, 2-14-5, 5-14-2, 3-14-4, 4-14-3, 1-13-7, 7-13-1, 2-13-6, 6-13-2, 3-13-5, 5-13-3, 4-13-4, 1-12-8, 8-12-1, 2-12-7, 7-12-2, 3-12-6, 6-12-3, 4-12-5, 5-12-4, 2-11-8, 8-11-2, 3-11-7, 7-11-3, 4-11-6, 6-11-4, 5-11-5, 1-20-1, 1-19-2, 2-19-1, 1-18-3, 3-18-1, 2-18-2, 1-17-4, 4-17-1, 2-17-3, 3-17-2, 1-16-5, 2-16-4, 4-16-2, 3-16-3, 1-15-6, 6-15-1, 2-15-5, 5-15-2, 3-15-4, 4-15-3, 1-14-7, 7-14-1, 2-14-6, 6-14-2, 3-14-5, 5-14-3, 4-14-4, 1-13-8, 8-13-1, 2-13-7, 7-13-2, 3-13-6, 6-13-3, 4-13-5, 5-13-4, 2-12-8, 8-12-2, 3-12-7, 7-12-3, 4-12-6, 6-12-4, 5-12-5, 3-11-8, 8-11-3, 4-11-7, 7-11-4, 5-11-6, 6-11-5, 1-21-1, 1-20-2, 2-20-1, 1-20-3, 3-19-1, 2-19-2, 1-18-4, 4-18-1, 2-18-3, 3-18-2, 1-17-5, 2-17-4, 4-17-2, 3-17-3, 1-16-6, 6-16-1, 2-16-5, 5-16-2, 3-16-4, 4-16-3, 1-15-7, 7-15-1, 2-15-6, 6-15-2, 3-15-5, 5-15-3, 4-15-4, 1-14-8, 8-14-1, 2-14-7, 7-14-2, 3-14-6, 6-14-3, 4-14-5, 5-14-4, 2-13-8, 8-13-2, 3-13-7, 7-13-3, 4-13-6, 6-13-4, 5-13-5, 1-12-10, 10-12-1, 2-12-9, 9-12-2, 3-12-8, 8-12-3, 4-12-7, 7-12-4, 5-12-6, 6-12-5, 4-11-8, 8-11-4, 5-11-7, 7-11-5, 6-11-6, 1-22-1, 1-21-2, 2-21-1, 1-21-3, 3-20-1, 2-20-2, 1-19-4, 4-19-1, 2-19-3, 3-19-2, 1-18-5, 2-18-4, 4-18-2, 3-18-3, 1-17-6, 6-17-1, 2-17-5, 5-17-2, 3-17-4, 4-17-3, 1-16-7, 7-16-1, 2-16-6, 6-16-2, 3-16-5, 5-16-3, 4-16-4, 1-15-8, 8-15-1, 2-15-7, 7-15-2, 3-15-6, 6-15-3, 4-15-5, 5-15-4, 2-14-8, 8-14-2, 3-14-7, 7-14-3, 4-14-6, 6-14-4, 5-14-5, 3-13-8, 8-13-3, 4-13-7, 7-13-4, 5-13-6, 6-13-5, 4-12-8, 8-12-4, 5-12-7, 7-12-5, 6-12-6, 5-11-8, 8-11-5, 6-11-7, or 7-11-6. In a particular embodiment, the gap-widened antisense oligonucleotides of the present invention have a 2-16-2, 3-14-3, or 4-12-4 wing-gap-wing motif.

Another aspect of the present invention is the use of a gap-widened antisense oligonucleotide 18-24 nucleotides in length comprising: a gap region having greater than 11 contiguous 2′-deoxyribonucleotides; and a first wing region and a second wing region flanking the gap region, wherein each of said first and second wing regions independently have 1 to 8 2′-O-(2-methoxyethyl)ribonucleotides, having an improved therapeutic index as compared to a corresponding 5-10-5 antisense oligonucleotide having a gap region of 10 contiguous 2′-deoxyribonucleotides and a first wing region and a second wing region flanking the gap region of 5 2′-O-(2-methoxyethyl)ribonucleotides in the manufacture of a medicament for the treatment of disorders and diseases related to target RNA levels. Another embodiment of the present invention is a pharmaceutical composition comprising a gap-widened antisense oligonucleotide 18-24 nucleotides in length comprising: a gap region having greater than 11 contiguous 2′-deoxyribonucleotides; and a first wing region and a second wing region flanking the gap region, wherein each of said first and second wing regions independently have 1 to 8 2′-O-(2-methoxyethyl)ribonucleotides, having an improved therapeutic index as compared to a corresponding 5-10-5 antisense oligonucleotide having a gap region of 10 contiguous 2′-deoxyribonucleotides and a first wing region and a second wing region flanking the gap region of 5 2′-O-(2-methoxyethyl)ribonucleotides and optionally a pharmaceutically acceptable carrier, diluent, enhancer or excipient. Another embodiment of the present invention is a gap-widened antisense oligonucleotide 18-24 nucleotides in length comprising: a gap region having greater than 11 contiguous 2′-deoxyribonucleotides; and a first wing region and a second wing region flanking the gap region, wherein each of said first and second wing regions independently have 1 to 8 2′-O-(2-methoxyethyl)ribonucleotides, having lower kidney accumulation as compared to a corresponding 5-10-5 antisense oligonucleotide having a gap region of 10 contiguous 2′-deoxyribonucleotides and a first wing region and a second wing region flanking the gap region of 5 2′-O-(2-methoxyethyl)ribonucleotides as measured by plasma protein binding capacity of said gap-widened antisense oligonucleotide. Also provided is a method of modulating gene expression in an animal comprising the step of contacting said animal with the pharmaceutical composition. Another embodiment is a method of modulating gene expression in an animal comprising the step of contacting said animal with a gap-widened antisense oligonucleotide of the invention wherein the accumulation of the gap-widened antisense oligonucleotide in the kidney is less compared to a corresponding 5-10-5 antisense oligonucleotide having a gap region of 10 contiguous 2′-deoxyribonucleotides and a first wing region and a second wing region flanking the gap region of 5 2′-O-(2-methoxyethyl)ribonucleotides. In one embodiment, the kidney accumulation is measured by plasma protein binding capacity of said gap-widened antisense oligonucleotide.

Another embodiment of the present invention is a method of reducing levels of a preselected RNA target in the liver of an animal comprising administering to said animal a chimeric antisense compound 11 to 80 nucleobases in length which is targeted to said preselected RNA target wherein said chimeric antisense compound comprises a first gap region consisting of at least 10 contiguous 2′-deoxynucleotides and a wing region which consists of from 1 to 4 contiguous nucleosides or nucleoside analogs which are not substrates for RNaseH. In particular embodiments, said first gap region consists of at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, or at least 18 contiguous 2′-deoxynucleotides. In one embodiment, the chimeric antisense compound comprises second wing region which consists of from 1 to 7 contiguous nucleosides or nucleoside analogs which are not substrates for RNase H, and wherein said gap region is located between said first wing region and said second wing region. In another embodiment, the chimeric antisense compound is a chimeric antisense oligonucleotide, and the nucleosides or nucleoside analog which is not a substrate for RNase H is a nucleotide having a 2′ modification of the sugar moiety. In one embodiment, the nucleotide having a 2′ modification of the sugar moiety is a 2′-O-methoxyethyl nucleotide. In some embodiments the compound is a 2-16-2 MOE gapmer, a 3-12-3 MOE gapmer, a 3-10-7 MOE gapmer or a 7-10-3 MOE gapmer. In one embodiment, the chimeric antisense oligonucleotide has at least one phosphorothioate backbone linkage.

Another embodiment of the present invention is a pharmaceutical composition for use in reducing levels of a preselected RNA target in the liver of an animal comprising a chimeric antisense compound targeted to said preselected RNA target, wherein said chimeric antisense compound comprises a first gap region consisting of at least 10 contiguous 2′-deoxynucleotides and a wing region which consists of from 1 to 4 contiguous nucleosides or nucleoside analogs which are not substrates for RNase H.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1. Depicts a comparison of in vivo effects of a 5-10-5 MOE gapmer (SEQ ID NO: 1) and the corresponding gap-widened 2-16-2 MOE gapmer (SEQ ID NO: 1).

FIG. 2. Depicts a comparison the persistence of target mRNA modulation of a 5-10-5 MOE gapmer (SEQ ID NO: 1) and the corresponding gap-widened 2-16-2 MOE gapmer (SEQ ID NO: 1).

FIG. 3. Depicts a comparison of in vitro effects of a 5-10-5 MOE gapmer and the corresponding gap-widened 2-16-2 MOE gapmer.

FIG. 4. Depicts a comparison of the concentrations of a 5-10-5 MOE gapmer and the corresponding gap-widened 2-16-2 MOE gapmer in liver and kidney tissues.

FIG. 5. Depicts a comparison of the in vitro effects of oligonucleotides having the same sequence (SEQ ID NO:3) but varied wing-gap-wing motifs.

FIG. 6. Depicts a comparison of the in vivo effects of oligonucleotides having the same sequence (SEQ ID NO: 3) but varied wing-gap-wing motifs.

DETAILED DESCRIPTION OF THE INVENTION

Certain gap sizes are optimal for in vivo efficacy of antisense compounds. Surprisingly, improved potency (3-10× improvement) in mouse or rat liver has been demonstrated for gap-widened antisense oligonucleotides compared to standard 5-10-5 MOE gapmer (for example, 2-16-2, 2-14-2, 3-12-3 gapmers) antisense oligonucleotides. This has been shown for several distinct antisense targets and this improved potency is not observed in cultured cells transfected with the same gap-widened antisense oligonucleotides. Thus the “gap-widened” motifs appear to convey some benefit to in vivo potency, particularly in the liver. It is demonstrated herein that chimeric antisense compounds having a gap of greater than eleven contiguous deoxynucleotides flanked by wing regions consisting of from 1 to 4 nucleotides which are not substrates for RNase H are particularly effective at reducing target RNA levels in vivo, particularly in the liver.

Therapeutic Index

Therapeutic index is a measure which relates the dose of a drug required to produce a specified effect to that which produces an undesired effect. In one embodiment, improved therapeutic index of a gap-widened antisense oligonucleotide is characterized by equal or increased potency and a reduction in tissue concentration. In another embodiment, improved therapeutic index of a gap-widened antisense oligonucleotide is characterized by increased potency and equal tissue concentrations as compared to a corresponding 5-10-5 antisense oligonucleotide. In another embodiment, improved therapeutic index of a gap-widened antisense oligonucleotide is characterized by increased potency and decreased toxicity as compared to a corresponding 5-10-5 antisense oligonucleotide. In another embodiment, improved therapeutic index of a gap-widened antisense oligonucleotide is characterized by comparable potency and decreased toxicity as compared to a corresponding 5-10-5 antisense oligonucleotide. In some embodiments, the toxicity is renal toxicity. In some embodiments, the toxicity is hepatic toxicity.

Indications

An embodiment of the present invention is a method of treating a disease or condition wherein a target RNA is associated with said disease or condition by administering a compound of the invention. Another embodiment of the present invention is a method of preventing or delaying the onset of a disease or condition wherein a target RNA is associated with said disease or condition by administering a compound of the invention. Diseases or conditions include metabolic and cardiovascular diseases or conditions. In some embodiments, the disease or condition is metabolic syndrome, diabetes, obesity, hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, Type 2 diabetes, diet-induced obesity, hyperglycemia, or insulin resistance. In one embodiment, the disease or condition is hepatic steatosis. In some embodiments, the steatosis is steatohepatitis or NASH. In some embodiments, the disease or condition is familial hypercholesterolemia, nonfamilial hypercholesterolemia, mixed dyslipidemia, dysbetalipoproteinemia, atherosclerosis, coronary artery disease, myocardial infarction, hypertension, carotid artery diseases, stroke, cerebrovascular disease, carotid artery disease, stroke, cerebrovascular disease, peripheral vascular disease, thrombosis, or arterial aneurism.

NAFLD and Metabolic Syndrome

The term “nonalcoholic fatty liver disease” (NAFLD) encompasses a disease spectrum ranging from simple triglyceride accumulation in hepatocytes (hepatic steatosis) to hepatic steatosis with inflammation (steatohepatitis), fibrosis, and cirrhosis. Nonalcoholic steatohepatitis (NASH) occurs from progression of NAFLD beyond deposition of triglycerides. A second-hit capable of inducing necrosis, inflammation, and fibrosis is required for development of NASH. Candidates for the second-hit can be grouped into broad categories: factors causing an increase in oxidative stress and factors promoting expression of proinflammatory cytokines. It has been suggested that increased liver triglycerides lead to increased oxidative stress in hepatocytes of animals and humans, indicating a potential cause-and-effect relationship between hepatic triglyceride accumulation, oxidative stress, and the progression of hepatic steatosis to NASH (Browning and Horton, J. Clin. Invest., 2004, 114, 147-152). Hypertriglyceridemia and hyperfattyacidemia can cause triglyceride accumulation in peripheral tissues (Shimamura et al., Biochem. Biophys. Res. Commun., 2004, 322, 1080-1085).

“Metabolic syndrome” is defined as a clustering of lipid and non-lipid cardiovascular risk factors of metabolic origin. It is closely linked to the generalized metabolic disorder known as insulin resistance. The National Cholesterol Education Program (NCEP) Adult Treatment Panel III (ATPIII) established criteria for diagnosis of metaolic syndrome when three or more of five risk determinants are present. The five risk determinants are abdominal obesity defined as waist circumference of greater than102 cm for men or greater than 88 cm for women, triglyceride levels greater than or equal to 150 mg/dL, HDL cholesterol levels of less than 40 mg/dL for men and less than 50 mg/dL for women, blood pressure greater than or equal to 130/85 mm Hg and fasting glucose levels greater than or equal to 110 mg/dL. These determinants can be readily measured in clinical practice (JAMA, 2001, 285, 2486-2497).

HbA1c

HbA1c is a stable minor hemoglobin variant formed in vivo via posttranslational modification by glucose, and it contains predominantly glycated NH2-terminal β-chains. There is a strong correlation between levels of HbA1c and the average blood glucose levels over the previous 3 months. Thus HbA1c is often viewed as the “gold standard” for measuring sustained blood glucose control (Bunn, H. F. et al., 1978, Science. 200, 21-7). HbA1c can be measured by ion-exchange HPLC or immunoassay; home blood collection and mailing kits for HbA1c measurement are now widely available. Serum fructosamine is another measure of stable glucose control and can be measured by a colorimetric method (Cobas Integra, Roche Diagnostics).

Cardiovascular Risk Profile

Conditions associated with risk of developing a cardiovascular disease include, but are not limited to, history of myocardial infarction, unstable angina, stable angina, coronary artery procedures (angioplasty or bypass surgery), evidence of clinically significant myocardial ischemia, noncoronary forms of atherosclerotic disease (peripheral arterial disease, abdominal aortic aneurysm, carotid artery disease), diabetes, cigarette smoking, hypertension, low HDL cholesterol, family history of premature CHD, obesity, physical inactivity, elevated triglyceride, or metabolic syndrome (Jama, 2001, 285, 2486-2497; Grundy et al., Circulation, 2004, 110, 227-239).

EXAMPLES Example 1 Oligonucleotide Sequences and Targets

TABLE 1 Oligonucleotide sequences (all are PS backbone) Modified nucleotides are shown in Bold (2′MOE unless otherwise indicated) and all cytosines are 5-methylcytosines SEQ ID ISIS No. Target Sequence NO Motif 116847 PTEN CTGCTAGCCTCTGGATTTGA 1 5-10-5 344266 PTEN CTGCTAGCCTCTGGATTTGA 1 2-16-2 141923 None (scrambled CCTTCCCTGAAGGTTCCTCC 2 5-10-5 117405 TRADD GCTCATACTCGTAGGCCA 3 4-10-4 325589 TRADD GCTCATACTCGTAGGCCA 3 5-8-5 325590 TRADD GCTCATACTCGTAGGCCA 3 6-6-6  29837 None (scrambled TCGATCTCCTTTTATGCCCG 4 5-10-5 325593 mTRADD CGCTCATACTCGTAGGCCAG 112 3-10-7 325594 TRADD CGCTCATACTCGTAGGCCAG 112 7-10-3 325584 TRADD CGCTCATACTCGTAGGCCAG 112 5-10-5 113715 PTP1B GCTCCTTCCACTGATCCTGC 113 5-10-5 344177 PTP1B GCTCCTTCCACTGATCCTGC 113 3-14-3 372350 GCCR TCTGTCTCTCCCATATACAG 5 2-16-2 372376 GCCR TGTTTCTGTCTCTCCCATAT 6 2-16-2 372331 GCCR CTTTTGTTTCTGTCTCTCCC 7 2-16-2 372341 GCCR ATCACTTTTGTTTCTGTCTC 8 2-16-2 352983 GCCR GTTTGCAATGCTTTCTTCCA 9 2-16-2 372365 GCCR TGAGGTTTGCAATGCTTTCT 10 2-16-2 372387 GCCR CTATTGAGGTTTGCAATGCT 11 2-16-2 372316 GCCR CGACCTATTGAGGTTTGCAA 12 2-16-2 372310 GCCR CTGGTCGACCTATTGAGGTT 13 2-16-2 372315 GCCR CTGTGGTATACAATTTCACA 14 2-16-2 372326 GCCR CTTTGGTCTGTGGTATACAA 15 2-16-2 372339 GCCR GTCAAAGGTGCTTTGGTCTG 16 2-16-2 372322 GCCR GGTTTAGTGTCCGGTAAAAT 17 2-16-2 372361 GCCR CTTTTTCTGTTTTCACTTGG 18 2-16-2 372308 GCCR TTCTCTTGCTTAATTACCCC 19 2-16-2 372304 GCCR CAGTTTCTCTTGCTTAATTA 20 2-16-2 352984 GCCR GCCCAGTTTCTCTTGCTTAA 21 2-16-2 372372 GCCR TTTATTACCAATTATATTTG 22 2-16-2 372327 GCCR ACATTTTATTACCAATTATA 23 2-16-2 372311 GCCR GCAGACATTTTATTACCAAT 24 2-16-2 372352 GCCR AATGGCAGACATTTTATTAC 25 2-16-2 372337 GCCR CAGAAATGGCAGACATTTTA 26 2-16-2 372323 GCCR TGAACAGAAATGGCAGACAT 27 2-16-2 372347 GCCR CCATGAACAGAAATGGCAGA 28 2-16-2 372383 GCCR CACACCATGAACAGAAATGG 29 2-16-2 372348 GCCR TACTCACACCATGAACAGAA 30 2-16-2 372363 GCCR GAGGTACTCACACCATGAAC 31 2-16-2 372334 GCCR TCCAGAGGTACTCACACCAT 32 2-16-2 372359 GCCR GTCCTCCAGAGGTACTCACA 33 2-16-2 372344 GCCR ATCTGTCCTCCAGAGGTACT 34 2-16-2 372307 GCCR GTACATCTGTCCTCCAGAGG 35 2-16-2 372370 GCCR AGTGGTACATCTGTCCTCCA 36 2-16-2 372374 GCCR TCATAGTGGTACATCTGTCC 37 2-16-2 372355 GCCR CATGTCATAGTGGTACATCT 38 2-16-2 372385 GCCR TATTCATGTCATAGTGGTAC 39 2-16-2 372319 GCCR GCTGTATTCATGTCATAGTG 40 2-16-2 372366 GCCR GGATGCTGTATTCATGTCAT 41 2-16-2 372330 GCCR AAAGGGATGCTGTATTCATG 42 2-16-2 372333 GCCR TGAGAAAGGGATGCTGTATT 43 2-16-2 372358 GCCR TGGTGGAATGACATTAAAAA 44 2-16-2 372381 GCCR GAATTGGTGGAATGACATTA 45 2-16-2 372377 GCCR GAGCTTACATCTGGTCTCAT 46 2-16-2 372309 GCCR AGGAGAGCTTACATCTGGTC 47 2-16-2 372388 GCCR ATGGAGGAGAGCTTACATCT 48 2-16-2 372321 GCCR CTGGATGGAGGAGAGCTTAC 49 2-16-2 372312 GCCR GAGCTGGATGGAGGAGAGCT 50 2-16-2 372324 GCCR TGTCCTTCCACTGCTCTTTT 51 2-16-2 372332 GCCR GTGCTGTCCTTCCACTGCTC 52 2-16-2 372335 GCCR AATTGTGCTGTCCTTCCACT 53 2-16-2 372342 GCCR AGGTAATTGTGCTGTCCTTC 54 2-16-2 372345 GCCR CGGCATGCTGGGCAGTTTTT 55 2-16-2 372356 GCCR ATAGCGGCATGCTGGGCAGT 56 2-16-2 372305 GCCR CGATAGCGGCATGCTGGGCA 57 2-16-2 372367 GCCR ATTCCAGCCTGAAGACATTT 58 2-16-2 372353 GCCR GTTCATTCCAGCCTGAAGAC 59 2-16-2 372364 GCCR TTCTTTGTTTTTCGAGCTTC 60 2-16-2 372340 GCCR TTTTTTCTTTGTTTTTCGAG 61 2-16-2 372369 GCCR CAGGAACTATTGTTTTGTTA 62 2-16-2 372378 GCCR TGCAGGAACTATTGTTTTGT 63 2-16-2 372317 GCCR GAGCTATCATATCCTGCATA 64 2-16-2 372351 GCCR AACAGAGCTATCATATCCTG 65 2-16-2 372389 GCCR CTGGAACAGAGCTATCATAT 66 2-16-2 372362 GCCR TTCACTGCTGCAATCACTTG 67 2-16-2 372328 GCCR CCATTTCACTGCTGCAATCA 68 2-16-2 372338 GCCR TTGCCCATTTCACTGCTGCA 69 2-16-2 372349 GCCR ATAATCAGATCAGGAGCAAA 70 2-16-2 372373 GCCR ATTAATAATCAGATCAGGAG 71 2-16-2 372360 GCCR GCTCATTAATAATCAGATCA 72 2-16-2 372384 GCCR CTCTGCTCATTAATAATCAG 73 2-16-2 372380 GCCR CATTCTCTGCTCATTAATAA 74 2-16-2 372320 GCCR AGCATGTGTTTACATTGGTC 75 2-16-2 372371 GCCR AAGGTTTTCATACAGAGATA 76 2-16-2 372382 GCCR CAGTAAGGTTTTCATACAGA 77 2-16-2 372306 GCCR GAAGCAGTAAGGTTTTCATA 78 2-16-2 372343 GCCR GAGAGAAGCAGTAAGGTTTT 79 2-16-2 372313 GCCR GCTTTTCCTAGCTCTTTGAT 80 2-16-2 372325 GCCR ATGGCTTTTCCTAGCTCTTT 81 2-16-2 372336 GCCR ATGGTCTTATCCAAAAATGT 82 2-16-2 372318 GCCR ACTCATGGTCTTATCCAAAA 83 2-16-2 372375 GCCR CAATACTCATGGTCTTATCC 84 2-16-2 372346 GCCR AATTCAATACTCATGGTCTT 85 2-16-2 372386 GCCR ATGATTTCAGCTAACATCTC 86 2-16-2 372354 GCCR GTGATGATTTCAGCTAACAT 87 2-16-2 372357 GCCR GAATATTTTGGTATCTGATT 88 2-16-2 372368 GCCR ATTTGAATATTTTGGTATCT 89 2-16-2 372379 GCCR TTCCATTTGAATATTTTGGT 90 2-16-2 372390 GCCR ATATTTCCATTTGAATATTT 91 2-16-2 372329 GCCR TTTTTGATATTTCCATTTGA 92 2-16-2 361132 GCCR TCTGTCTCTCCCATATACAG 5 5-10-5 361133 GCCR TGTTTCTGTCTCTCCCATAT 6 5-10-5 361134 GCCR CTTTTGTTTCTGTCTCTCCC 7 5-10-5 361135 GCCR ATCACTTTTGTTTCTGTCTC 8 5-10-5 180272 GCCR GTTTGCAATGCTTTCTTCCA 9 5-10-5 345188 GCCR TGAGGTTTGCAATGCTTTCT 10 5-10-5 361136 GCCR CTATTGAGGTTTGCAATGCT 11 5-10-5 361137 GCCR CGACCTATTGAGGTTTGCAA 12 5-10-5 180274 GCCR CTGGTCGACCTATTGAGGTT 13 5-10-5 180275 GCCR CTGTGGTATACAATTTCACA 14 5-10-5 180276 GCCR CTTTGGTCTGTGGTATACAA 15 5-10-5 345198 GCCR GTCAAAGGTGCTTTGGTCTG 16 5-10-5 180279 GCCR GGTTTAGTGTCCGGTAAAAT 17 5-10-5 361138 GCCR CTTTTTCTGTTTTCACTTGG 18 5-10-5 180280 GCCR TTCTCTTGCTTAATTACCCC 19 5-10-5 345218 GCCR CAGTTTCTCTTGCTTAATTA 20 5-10-5 180281 GCCR GCCCAGTTTCTCTTGCTTAA 21 5-10-5 361139 GCCR TTTATTACCAATTATATTTG 22 5-10-5 361140 GCCR ACATTTTATTACCAATTATA 23 5-10-5 361141 GCCR GCAGACATTTTATTACCAAT 24 5-10-5 361142 GCCR AATGGCAGACATTTTATTAC 25 5-10-5 361143 GCCR CAGAAATGGCAGACATTTTA 26 5-10-5 361144 GCCR TGAACAGAAATGGCAGACAT 27 5-10-5 180283 GCCR CCATGAACAGAAATGGCAGA 28 5-10-5 361145 GCCR CACACCATGAACAGAAATGG 29 5-10-5 361146 GCCR TACTCACACCATGAACAGAA 30 5-10-5 361147 GCCR GAGGTACTCACACCATGAAC 31 5-10-5 361148 GCCR TCCAGAGGTACTCACACCAT 32 5-10-5 361149 GCCR GTCCTCCAGAGGTACTCACA 33 5-10-5 361150 GCCR ATCTGTCCTCCAGAGGTACT 34 5-10-5 361151 GCCR GTACATCTGTCCTCCAGAGG 35 5-10-5 361152 GCCR AGTGGTACATCTGTCCTCCA 36 5-10-5 361153 GCCR TCATAGTGGTACATCTGTCC 37 5-10-5 361154 GCCR CATGTCATAGTGGTACATCT 38 5-10-5 361155 GCCR TATTCATGTCATAGTGGTAC 39 5-10-5 361156 GCCR GCTGTATTCATGTCATAGTG 40 5-10-5 361157 GCCR GGATGCTGTATTCATGTCAT 41 5-10-5 361158 GCCR AAAGGGATGCTGTATTCATG 42 5-10-5 180288 GCCR TGAGAAAGGGATGCTGTATT 43 5-10-5 180289 GCCR TGGTGGAATGACATTAAAAA 44 5-10-5 361159 GCCR GAATTGGTGGAATGACATTA 45 5-10-5 361160 GCCR GAGCTTACATCTGGTCTCAT 46 5-10-5 361161 GCCR AGGAGAGCTTACATCTGGTC 47 5-10-5 361162 GCCR ATGGAGGAGAGCTTACATCT 48 5-10-5 361163 GCCR CTGGATGGAGGAGAGCTTAC 49 5-10-5 361164 GCCR GAGCTGGATGGAGGAGAGCT 50 5-10-5 361165 GCCR TGTCCTTCCACTGCTCTTTT 51 5-10-5 361166 GCCR GTGCTGTCCTTCCACTGCTC 52 5-10-5 361167 GCCR AATTGTGCTGTCCTTCCACT 53 5-10-5 361168 GCCR AGGTAATTGTGCTGTCCTTC 54 5-10-5 361169 GCCR CGGCATGCTGGGCAGTTTTT 55 5-10-5 361170 GCCR ATAGCGGCATGCTGGGCAGT 56 5-10-5 361171 GCCR CGATAGCGGCATGCTGGGCA 57 5-10-5 361172 GCCR ATTCCAGCCTGAAGACATTT 58 5-10-5 361173 GCCR GTTCATTCCAGCCTGAAGAC 59 5-10-5 361174 GCCR TTCTTTGTTTTTCGAGCTTC 60 5-10-5 361175 GCCR TTTTTTCTTTGTTTTTCGAG 61 5-10-5 180297 GCCR CAGGAACTATTGTTTTGTTA 62 5-10-5 361176 GCCR TGCAGGAACTATTGTTTTGT 63 5-10-5 361177 GCCR GAGCTATCATATCCTGCATA 64 5-10-5 361178 GCCR AACAGAGCTATCATATCCTG 65 5-10-5 361179 GCCR CTGGAACAGAGCTATCATAT 66 5-10-5 361180 GCCR TTCACTGCTGCAATCACTTG 67 5-10-5 361181 GCCR CCATTTCACTGCTGCAATCA 68 5-10-5 361182 GCCR TTGCCCATTTCACTGCTGCA 69 5-10-5 361183 GCCR ATAATCAGATCAGGAGCAAA 70 5-10-5 361184 GCCR ATTAATAATCAGATCAGGAG 71 5-10-5 361185 GCCR GCTCATTAATAATCAGATCA 72 5-10-5 361186 GCCR CTCTGCTCATTAATAATCAG 73 5-10-5 180302 GCCR CATTCTCTGCTCATTAATAA 74 5-10-5 180304 GCCR AGCATGTGTTTACATTGGTC 75 5-10-5 361187 GCCR AAGGTTTTCATACAGAGATA 76 5-10-5 361188 GCCR CAGTAAGGTTTTCATACAGA 77 5-10-5 361189 GCCR GAAGCAGTAAGGTTTTCATA 78 5-10-5 180307 GCCR GAGAGAAGCAGTAAGGTTTT 79 5-10-5 361190 GCCR GCTTTTCCTAGCTCTTTGAT 80 5-10-5 361191 GCCR ATGGCTTTTCCTAGCTCTTT 81 5-10-5 361192 GCCR ATGGTCTTATCCAAAAATGT 82 5-10-5 361193 GCCR ACTCATGGTCTTATCCAAAA 83 5-10-5 361194 GCCR CAATACTCATGGTCTTATCC 84 5-10-5 361195 GCCR AATTCAATACTCATGGTCTT 85 5-10-5 361196 GCCR ATGATTTCAGCTAACATCTC 86 5-10-5 180311 GCCR GTGATGATTTCAGCTAACAT 87 5-10-5 361197 GCCR GAATATTTTGGTATCTGATT 88 5-10-5 361198 GCCR ATTTGAATATTTTGGTATCT 89 5-10-5 361199 GCCR TTCCATTTGAATATTTTGGT 90 5-10-5 361200 GCCR ATATTTCCATTTGAATATTT 91 5-10-5 361202 GCCR TTTTTGATATTTCCATTTGA 92 5-10-5 310457 GCGR GCACTTTGTGGTGCCAAGGC 93 5-10-5 325448 GCGR GCACTTTGTGGTGCCAAGGC 93 2-16-2 325568 GCGR GCACTTTGTGGTGCCAAGGC 93 3-14-3 356171 GCGR GCACTTTGTGGTACCAAGGT 94 5-10-5 357368 GCGR GCACTTTGTGGTACCAAGGT 94 Uniform deoxy 357369 GCGR GCACTTTGTGGTACCAAGGT 94 1-18-1 357370 GCGR GCACTTTGTGGTACCAAGGT 94 1-17-2 357371 GCGR GCACTTTGTGGTACCAAGGT 94 2-16-2 357372 GCGR GCACTTTGTGGTACCAAGGT 94 3-14-3 357373 GCGR GCACTTTGTGGTACCAAGGT 94 4-12-4 217328 DGAT2 GCATTGCCACTCCCATTCTT 95 5-10-5 334177 DGAT2 AGGACCCCGGAGTAGGCGGC 96 5-10-5 366710 DGAT2 GACCTATTGAGCCAGGTGAC 97 5-10-5 366714 DGAT2 GTAGCTGCTTTTCCACCTTG 98 5-10-5 370727 DGAT2 AGCTGCTTTTCCACCTTGGA 99 2-16-2 370747 DGAT2 TGGAGCTCAGAGACTCAGCC 100 2-16-2 370784 DGAT2 GCTGCATCCATGTCATCAGC 101 2-16-2

TABLE 2 Target sequences Target name Synonyms Species GENBANK Accession No or description SEQ ID NO PTEN MMAC1; TEP1; TGF beta regulated and mouse U92437.1 103 epithelial cell-enriched phosphatase; mutated in multiple advanced cancers 1; phosphatase and tensin homologue; putative protein tyrosine phosphatase TRADD TNF receptor 1 associated protein; mouse consensus sequence built from mouse ESTs: 104 TNFRSF1A-associated via death domain; aa013629, aa914725, aa013699, aa122508, aa881900, Tumor necrosis factor receptor associated aa423244, aa930854, w13708, aa201054, ai122320, death domain aa611848, aa546092, and aa939422 GCCR nuclear receptor subfamily 3, group C, human NM_000176.1 105 member 1; GR; GRL; NR3C1; rat NM_012576.1 106 glucocorticoid receptor; nuclear receptor mouse NM_008173.1 107 subfamily 3, group C, member 1 GCGR glucagon receptor; GR human NM_000160.1 108 rat M96674.1 109 DGAT2 ACYL-CoA: DIACYLGLYCEROL human NM_032564.2 110 ACYLTRANSFERASE 2; diacylglycerol rat the complement of nucleotides 15333000 to 111 acyltransferase 2; DIACYLGLYCEROL O- 15365000 of GENBANK ® accession number ACYLTRANSFERASE 2; GS1999full; NW_047561.1 LOC84649 PTP1B PTP-1B; PTPN1; RKPTP; protein tyrosine human M31724.1 114 phosphatase; protein tyrosine phosphatase 1B; protein tyrosine phosphatase, non- receptor type 1

Example 2 Assaying Modulation of Expression

Modulation of target RNA expression can be assayed in a variety of ways known in the art. GCCR mRNA levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or real-time PCR. RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA by methods known in the art. Methods of RNA isolation are taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 1, pp. 4.1.1-4.2.9 and 4.5.1-4.5.3, John Wiley & Sons, Inc., 1993.

Northern blot analysis is routine in the art and is taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 1, pp. 4.2.1-4.2.9, John Wiley & Sons, Inc., 1996. Real-time quantitative (PCR) can be conveniently accomplished using the commercially available ABI PRISM™ 7700 Sequence Detection System, available from PE-Applied Biosystems, Foster City, Calif. and used according to manufacturer's instructions.

Levels of proteins encoded by a target RNA can be quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), ELISA or fluorescence-activated cell sorting (FACS). Antibodies directed to a protein encoded by a target RNA can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, Mich.), or can be prepared via conventional antibody generation methods. Methods for preparation of polyclonal antisera are taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 11.12.1-11.12.9, John Wiley & Sons, Inc., 1997. Preparation of monoclonal antibodies is taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 11.4.1-11.11.5, John Wiley & Sons, Inc., 1997.

Immunoprecipitation methods are standard in the art and can be found at, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 10.16.1-10.16.11, John Wiley & Sons, Inc., 1998. Western blot (immunoblot) analysis is standard in the art and can be found at, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 10.8.1-10.8.21, John Wiley & Sons, Inc., 1997. Enzyme-linked immunosorbent assays (ELISA) are standard in the art and can be found at, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 11.2.1-11.2.22, John Wiley & Sons, Inc., 1991.

The effect of oligomeric compounds of the present invention on target nucleic acid expression can be tested in any of a variety of cell types provided that the target nucleic acid is present at measurable levels. The effect of oligomeric compounds of the present invention on target nucleic acid expression can be routinely determined using, for example, PCR or Northern blot analysis. Cell lines are derived from both normal tissues and cell types and from cells associated with various disorders (e.g. hyperproliferative disorders). Cell lines derived from multiple tissues and species can be obtained from American Type Culture Collection (ATCC, Manassas, Va.), the Japanese Cancer Research Resources Bank (Tokyo, Japan), or the Centre for Applied Microbiology and Research (Wiltshire, United Kingdom).

Primary cells, or those cells which are isolated from an animal and not subjected to continuous culture, can be prepared according to methods known in the art or obtained from various commercial suppliers. Additionally, primary cells include those obtained from donor human subjects in a clinical setting (i.e. blood donors, surgical patients).

Cell Types

The effects of oligomeric compounds on target nucleic acid expression were tested in the following cell types:

b.END Cells:

The mouse brain endothelial cell line b.END was obtained from Dr. Werner Risau at the Max Plank Institute (Bad Nauheim, Germany). b.END cells were routinely cultured in DMEM, high glucose (Invitrogen Life Technologies, Carlsbad, Calif.) supplemented with 10% fetal bovine serum (Invitrogen Life Technologies, Carlsbad, Calif.). Cells were routinely passaged by trypsinization and dilution when they reached approximately 90% confluence. Cells were seeded into 96-well plates (Falcon-Primaria #3872) at a density of approximately 3000 cells/well for use in oligomeric compound transfection experiments.

HepG2 Cells:

The human hepatoblastoma cell line HepG2 was obtained from the American Type Culture Collection (Manassas, Va.). HepG2 cells were routinely cultured in Eagle's MEM supplemented with 10% fetal bovine serum, 1 mM non-essential amino acids, and 1 mM sodium pyruvate (Invitrogen Life Technologies, Carlsbad, Calif.). Cells were routinely passaged by trypsinization and dilution when they reached approximately 90% confluence. Multiwell culture plates are prepared for cell culture by coating with a 1:100 dilution of type 1 rat tail collagen (BD Biosciences, Bedford, Mass.) in phosphate-buffered saline. The collagen-containing plates were incubated at 37° C. for approximately 1 hour, after which the collagen was removed and the wells were washed twice with phosphate-buffered saline. Cells were seeded into 96-well plates (Falcon-Primaria #353872, BD Biosciences, Bedford, Mass.) at a density of approximately 8,000 cells/well for use in oligomeric compound transfection experiments.

Primary Rat Hepatocytes:

Primary rat hepatocytes are prepared from Sprague-Dawley rats purchased from Charles River Labs (Wilmington, Mass.) and are routinely cultured in DMEM, high glucose (Invitrogen Life Technologies, Carlsbad, Calif.) supplemented with 10% fetal bovine serum (Invitrogen Life Technologies, Carlsbad, Calif.), 100 units per mL penicillin, and 100 μg/mL streptomycin (Invitrogen Life Technologies, Carlsbad, Calif.). Cells are seeded into 96-well plates (Falcon-Primaria #353872, BD Biosciences, Bedford, Mass.) at a density of approximately 4,000-6,000 cells/well treatment with the oligomeric compounds of the invention.

Treatment with Oligomeric Compounds

When cells reached appropriate confluency, they were treated with oligonucleotide using a transfection method as described. Other suitable transfection reagents known in the art include, but are not limited to, LIPOFECTAMINE™, CYTOFECTIN™, OLIGOFECTAMINE™, and FUGENE™. Other suitable transfection methods known in the art include, but are not limited to, electroporation.

LIPOFECTIN™

When cells reach 65-75% confluency, they are treated with oligonucleotide. Oligonucleotide is mixed with UPOFECTIN™ Invitrogen Life Technologies, Carlsbad, Calif.) in Opti-MEM™-1 reduced serum medium (Invitrogen Life Technologies, Carlsbad, Calif.) to achieve the desired concentration of oligonucleotide and a LIPOFECTIN™ concentration of 2.5 or 3 μg/mL per 100 nM oligonucleotide. This transfection mixture is incubated at room temperature for approximately 0.5 hours. For cells grown in 96-well plates, wells are washed once with 100 μL OPTI-MEM™-1 and then treated with 130 μL of the transfection mixture. Cells grown in 24-well plates or other standard tissue culture plates are treated similarly, using appropriate volumes of medium and oligonucleotide. Cells are treated and data are obtained in duplicate or triplicate. After approximately 4-7 hours of treatment at 37° C., the medium containing the transfection mixture is replaced with fresh culture medium. Cells are harvested 16-24 hours after oligonucleotide treatment.

Example 3 Real-Time Quantitative PCR Analysis of GCCR mRNA Levels

Quantitation of GCCR mRNA levels was accomplished by real-time quantitative PCR using the ABI PRISM™ 7600, 7700, or 7900 Sequence Detection System (PE-Applied Biosystems, Foster City, Calif.) according to manufacturer's instructions.

Gene target quantities obtained by RT, real-time PCR were normalized using either the expression level of GAPDH, a gene whose expression is constant, or by quantifying total RNA using RiboGreen™ (Molecular Probes, Inc. Eugene, Oreg.). Total RNA was quantified using RiboGreen™ RNA quantification reagent (Molecular Probes, Inc. Eugene, Oreg.). 170 μL of RiboGreen™ working reagent (RiboGreen™ reagent diluted 1:350 in 10 mM Tris-HCl, 1 mM EDTA, pH 7.5) was pipetted into a 96-well plate containing 30 μL purified cellular RNA. The plate was read in a CytoFluor 4000 (PE Applied Biosystems) with excitation at 485 nm and emission at 530 nm.

GAPDH expression was quantified by RT, real-time PCR, either simultaneously with the quantification of the target or separately. For measurement simultaneous with measurement of target levels, primer-probe sets specific to the target gene being measured were evaluated for their ability to be “multiplexed” with a GAPDH amplification reaction prior to quantitative PCR analysis. Multiplexing refers to the detection of multiple DNA species, in this case the target and endogenous GAPDH control, in a single tube, which requires that the primer-probe set for GAPDH does not interfere with amplification of the target.

Probes and primers for use in real-time PCR were designed to hybridize to target-specific sequences. Methods of primer and probe design are known in the art. Design of primers and probes for use in real-time PCR can be carried out using commercially available software, for example Primer Express®, PE Applied Biosystems, Foster City, Calif. The target-specific PCR probes have FAM covalently linked to the 5′ end and TAMRA or MGB covalently linked to the 3′ end, where FAM is the fluorescent dye and TAMRA or MGB is the quencher dye.

After isolation, the RNA is subjected to sequential reverse transcriptase (RT) reaction and real-time PCR, both of which are performed in the same well. RT and PCR reagents were obtained from Invitrogen Life Technologies (Carlsbad, Calif.). RT, real-time PCR was carried out in the same by adding 20 μL PCR cocktail (2.5× PCR buffer minus MgCl2, 6.6 mM MgCl2, 375 μM each of dATP, dCTP, dCTP and dGTP, 375 nM each of forward primer and reverse primer, 125 nM of probe, 4 Units RNAse inhibitor, 1.25 Units PLATINUM® Taq, 5 Units MuLV reverse transcriptase, and 2.5×ROX dye) to 96-well plates containing 30 μL total RNA solution (20-200 ng). The RT reaction was carried out by incubation for 30 minutes at 48° C. Following a 10 minute incubation at 95° C. to activate the PLATINUM® Taq, 40 cycles of a two-step PCR protocol were carried out: 95° C. for 15 seconds (denaturation) followed by 60° C. for 1.5 minutes (annealing/extension).

Example 4 Increased Potency of ISIS 344266 (2-16-2) In Vivo Compared to 5-10-5 Compound is not due to Enhanced Oligonucleotide Accumulation

Mice were dosed with ISIS 116847 (SEQ ID NO: 1) or ISIS 344266 (SEQ ID NO: 1) at 6, 3, 1.5 or 0.75 micromol/kg (approx 40, 20, 10 or 5 mg per kg), twice a week for three weeks and sacrificed 48 hours after the last dose was given. The left panel of FIG. 1 is a graph showing percent reduction of target RNA in liver following administration of saline, ISIS 141923 (negative unrelated control oligonucleotide, incorporated herein as SEQ ID NO: 2), ISIS 116847 at four concentrations, or ISIS 344266 at four concentrations. Both ISIS 116847 and ISIS 344266 are targeted to mouse PTEN (GENBANK® Accession No: U92437.1, herein incorporated as SEQ ID NO: 103), and are cross-species oligonucleotides with perfect complementary to human, rat, and rabbit PTEN. Neither saline nor negative control ISIS 141923 (6 micromoles/kg) reduced PTEN RNA levels. ISIS 116847 reduced PTEN RNA levels by approximately 21%, 44%, 64% and 81% at doses of 0.75, 1.5, 3 and 6 micromol/kg, respectively. ISIS 344266, the gap-widened antisense oligonucleotide, reduced PTEN RNA levels by approximately 54%, 79%, 88% and 91% at doses of 0.75, 1.5, 3 and 6 micromol/kg, respectively. A corresponding reduction of PTEN protein was demonstrated by Western blot as shown in the right panel of FIG. 1.

The ID50 (dose resulting in 50% reduction of PTEN RNA) calculated from these results was 1.9 micromol/kg for 116847 and 0.63 micromol/kg for 344266. The IC50 for ISIS 116847 was also over three-fold that of ISIS 344266. These results indicate that the gap-widened antisense oligonucleotide is three-fold more potent than the 5-10-5 compound of equivalent sequence.

ISIS 344266 (2-16-2) supports similar persistence of action compared to ISIS 116847 (5-10-5). Mice were treated as described above with ISIS 344266 (1.5 or 6 micromol/kg) or ISIS 116847 (6 micromol/kg), or with saline. PTEN RNA levels were measured in mouse liver at days 1, 7, 14 and 28. As shown in FIG. 2, the two compounds show similar durability of reduction of PTEN RNA levels, and even after 28 days the PTEN RNA levels in antisense-treated animals (either 116847 or 344266) had not returned to control levels.

The advantage conveyed by the gap-widened antisense oligonucleotides of the present invention for target reduction in vivo is surprising because it is not observed in vitro. An in vitro comparison of the same PTEN oligonucleotides, ISIS 116847 (5-10-5) and ISIS 344266 (2-16-2) was performed in cultured mouse bEND cells. Cells were transfected with oligonucleotide at doses of 0.1 nM, 0.3 nM, 0.9 nM, 2.7 nM, 8.1 nM and 24.3 nM in the presence of 3 microgram/ml LIPOFECTIN. Reduction of target expression was assayed by quantitative RT real-time PCR as described herein. FIG. 3 shows that the 5-10-5 gapmer was less potent than the 2-16-2 gapmer. The IC50 for reduction of PTEN RNA by the 5-10-5 gapmer (ISIS 116847) was 3.4 nM and 6.2 nM for the 2-16-2 gapmer (ISIS 344266). Thus the advantage conveyed by the gap-widened antisense oligonucleotides for target reduction in liver is not observed in cultured cells.

The enhanced potency of the gap-widened (2-16-2) PTEN antisense oligonucleotide in liver is not due to increased concentrations in liver compared to the 5-10-5 gapmer. Oligonucleotide concentration in kidney and liver tissue from mice treated as described above with ISIS 116847 or ISIS 344266 were determined. Methods to determine oligonucleotide concentration in tissues are known in the art (Geary et al., Anal Biochem, 1999, 274, 241-248). Oligonucleotide concentrations (micrograms/gram) in mouse liver and kidney were determined. As shown in FIG. 4, there was consistently less ISIS 344266 than ISIS 116847 in liver at every oligonucleotide dosage. The same is true for kidney although overall concentrations of both compounds were lower in kidney. Thus, the enhanced potency of the gap-widened antisense oligonucleotide (2-16-2 chimera) in the liver is not due to enhanced accumulation of compound in the liver.

Serum transaminases (AST/ALT) were higher for mice treated with 2-16-2 compound (ISIS 344266) than for those treated with ISIS 116847. However, because ISIS 344266 is more potent (active at lower doses), the therapeutic window for the two compounds is roughly comparable.

Example 5 Effect of Gap Size on In Vitro and In Vivo Potency

A series of MOE gapmers (2-14-2 through 6-6-6) were designed to target mouse TRADD (consensus sequence built from mouse ESTs: aa013629,aa914725,aa013699, aa122508, aa881900, aa423244, aa930854, w13708, aa201054, ai122320, aa611848, aa546092, and aa939422, incoporated herein as SEQ ID NO: 104). As shown in Table 2, a series of 18 mer chimeric antisense oligonucleotides were synthesized, all having the same sequence (GCTCATACTCGTAGGCCA, incorporated herein as SEQ ID NO: 3). Plain text indicates a deoxynucleotide, and nucleobases designated with bold, underlined text are 2′-O-(2-methoxyethyl)nucleotides. Internucleoside linkages are phosphorothioate throughout, and all cytosines are 5-methylcytosines. Indicated in Table 2 is the “motif' of each compound indicative of chemically distinct regions comprising the oligonucleotide.

TABLE 2 Antisense oligonucleotides targeting mouse TRADD ISIS Number Chemistry Motif ISIS 325589 GCTCATACTCGTAGGCCA 5-8-5 ISIS 117405 GCTCATACTCGTAGGCCA 4-10-4 ISIS 325588 GCTCATACTCGTAGGCCA 3-12-3 ISIS 325587 GCTCATACTCGTAGGCCA 2-14-2 ISIS 325590 GCTCATACTCGTAGGCCA 6-6-6

The compounds were tested in vitro in mouse bEND cells at concentrations of 0.1 nM, 0.5 nM, 2.5 nM, 12.5 nM and 62.5 nM for their ability to reduce target mRNA levels using real-time PCR as described herein. As shown in FIG. 5, in vitro IC50s for these compounds were 9.2 nM for the 5-8-5 gapmer (ISIS 325589), 11 nM for the 4-10-4 gapmer (ISIS 117405), 19 nM for the 3-12-3 gapmer (ISIS 325588), 49 nM for the 2-142 gapmer (ISIS 325587) and 82 nM for the 6-6-6 gapmer (ISIS 325590). Thus in this in vitro experiment, larger gaps did not appear to convey added potency.

When these compounds were tested in vivo, a different rank order potency was observed. Mice were treated with TRADD gapmer oligos (described above) ranging from 2-14-2 chimeras to 6-6-6 chimeras, each at doses of 1.56 micromole/kg, 3.12 micromol/kg and 6.24 micromol/kg. The negative control was ISIS 29837 (SEQ ID NO: 4) and animals treated with saline alone served as the control group to which data were normalized. As shown in FIG. 6, potency in liver increased with increasing gap size (from 6 to 14 contiguous deoxynucleotides). In a subsequent experiment (not shown) the 2-14-2 compound was approximately two-fold better than the 4-10-4 compound.

The effect of these gapmer compounds on mouse body weight, liver weight and spleen weights was compared and no meaningful differences were seen. Mice gained weight at roughly the same rate (indicating general good health) and liver and spleen weights were comparable to saline in all the treatment groups.

Example 6 Antisense Inhibition of Human GCCR Expression by 5-10-5 Gapmers or 2-16-2 Gapmers In Vitro

A series of oligomeric compounds was designed to target different regions of human GCCR, using published sequences (GENBANK® accession no: NM000176.1, incoporated herein as SEQ ID NO: 105). The compounds are shown in Table 3. All compounds in Table 3 are chimeric oligonucleotides (”gapmers“) 20 nucleotides in length, composed of a central “gap” region consisting of 10 2′-deoxynucleotides, which is flanked on both sides (5′ and 3′) by five-nucleotide “wings”. The wings are composed of 2′-O-(2-methoxyethyl)nucleotides, also known as 2′-MOE nucleotides. The internucleoside (backbone) linkages are phosphorothioate throughout the oligonucleotide. All cytidine residues are 5-methylcytidines. Shown in Table 3 is the sequence of the oligonucleotide, and the target site which is the first (5′ most) position on the target sequence to which the compound binds. The compounds were analyzed for their effect on gene target mRNA levels by quantitative real-time PCR as described in other examples herein, using a primer-probe set designed to hybridize to human GCCR.

Data are averages from three experiments in which HepG2 cells were treated with 50 nM of the disclosed oligomeric compounds using LIPOFECTIN™. A reduction in expression is expressed as percent inhibition in Table 3. If present, “N.D.” indicates “not determined”. The target regions to which these oligomeric compounds are inhibitory are herein referred to as “validated target segments.”

TABLE 3 Inhibition of human GCCR mRNA levels by 5-10-5 gapmers ISIS Target SEQ No of SEQ ID Target % Inhib ID 5-10-5 NO Site Sequence w/5-10-5 NO 361132 105  394 TCTGTCTCTCCCATATACAG 65  5 361133 105  398 TGTTTCTGTCTCTCCCATAT 56  6 361134 105  402 CTTTTGTTTCTGTCTCTCCC 60  7 361135 105  406 ATCACTTTTGTTTCTGTCTC 80  8 180272 105  497 GTTTGCAATGCTTTCTTCCA 74  9 345188 105  501 TGAGGTTTGCAATGCTTTCT 71 10 361136 105  505 CTATTGAGGTTTGCAATGCT 10 11 361137 105  509 CGACCTATTGAGGTTTGCAA 80 12 180274 105  514 CTGGTCGACCTATTGAGGTT 68 13 180275 105  672 CTGTGGTATACAATTTCACA 44 14 180276 105  679 CTTTGGTCTGTGGTATACAA 78 15 345198 105  689 GTCAAAGGTGCTTTGGTCTG 79 16 180279 105  877 GGTTTAGTGTCCGGTAAAAT 60 17 361138 105  954 CTTTTTCTGTTTTCACTTGG 70 18 180280 105 1000 TTCTCTTGCTTAATTACCCC 77 19 345218 105 1004 CAGTTTCTCTTGCTTAATTA 67 20 180281 105 1007 GCCCAGTTTCTCTTGCTTAA 74 21 361139 105 1058 TTTATTACCAATTATATTTG  0 22 361140 105 1062 ACATTTTATTACCAATTATA 35 23 361141 105 1066 GCAGACATTTTATTACCAAT 78 24 361142 105 1070 AATGGCAGACATTTTATTAC 40 25 361143 105 1074 CAGAAATGGCAGACATTTTA 63 26 361144 105 1078 TGAACAGAAATGGCAGACAT 61 27 180283 105 1081 CCATGAACAGAAATGGCAGA 69 28 361145 105 1085 CACACCATGAACAGAAATGG 30 29 361146 105 1089 TACTCACACCATGAACAGAA 60 30 361147 105 1093 GAGGTACTCACACCATGAAC 71 31 361148 105 1097 TCCAGAGGTACTCACACCAT 75 32 361149 105 1101 GTCCTCCAGAGGTACTCACA 69 33 361150 105 1105 ATCTGTCCTCCAGAGGTACT 53 34 361151 105 1109 GTACATCTGTCCTCCAGAGG 75 35 361152 105 1113 AGTGGTACATCTGTCCTCCA 62 36 361153 105 1117 TCATAGTGGTACATCTGTCC 52 37 361154 105 1121 CATGTCATAGTGGTACATCT 57 38 361155 105 1125 TATTCATGTCATAGTGGTAC 41 39 361156 105 1129 GCTGTATTCATGTCATAGTG 67 40 361157 105 1133 GGATGCTGTATTCATGTCAT 67 41 361158 105 1137 AAAGGGATGCTGTATTCATG 45 42 180288 105 1141 TGAGAAAGGGATGCTGTATT 62 43 180289 105 1181 TGGTGGAATGACATTAAAAA 54 44 361159 105 1185 GAATTGGTGGAATGACATTA 24 45 361160 105 1324 GAGCTTACATCTGGTCTCAT 59 46 361161 105 1328 AGGAGAGCTTACATCTGGTC 65 47 361162 105 1332 ATGGAGGAGAGCTTACATCT 18 48 361163 105 1336 CTGGATGGAGGAGAGCTTAC 50 49 361164 105 1339 GAGCTGGATGGAGGAGAGCT 49 50 361165 105 1468 TGTCCTTCCACTGCTCTTTT 61 51 361166 105 1472 GTGCTGTCCTTCCACTGCTC 65 52 361167 105 1476 AATTGTGCTGTCCTTCCACT 62 53 361168 105 1480 AGGTAATTGTGCTGTCCTTC 52 54 361169 105 1543 CGGCATGCTGGGCAGTTTTT 78 55 361170 105 1547 ATAGCGGCATGCTGGGCAGT 58 56 361171 105 1549 CGATAGCGGCATGCTGGGCA 65 57 361172 105 1570 ATTCCAGCCTGAAGACATTT 24 58 361173 105 1574 GTTCATTCCAGCCTGAAGAC 52 59 361174 105 1597 TTCTTTGTTTTTCGAGCTTC 62 60 361175 105 1601 TTTTTTCTTTGTTTTTCGAG 48 61 180297 105 1680 CAGGAACTATTGTTTTGTTA 33 62 361176 105 1682 TGCAGGAACTATTGTTTTGT 46 63 361177 105 1765 GAGCTATCATATCCTGCATA 71 64 361178 105 1769 AACAGAGCTATCATATCCTG 51 65 361179 105 1773 CTGGAACAGAGCTATCATAT 67 66 361180 105 1840 TTCACTGCTGCAATCACTTG 52 67 361181 105 1844 CCATTTCACTGCTGCAATCA 55 68 361182 105 1848 TTGCCCATTTCACTGCTGCA 70 69 361183 105 1999 ATAATCAGATCAGGAGCAAA 36 70 361184 105 2003 ATTAATAATCAGATCAGGAG 10 71 361185 105 2007 GCTCATTAATAATCAGATCA 43 72 361186 105 2011 CTCTGCTCATTAATAATCAG  0 73 180302 105 2015 CATTCTCTGCTCATTAATAA 23 74 180304 105 2053 AGCATGTGTTTACATTGGTC 73 75 361187 105 2119 AAGGTTTTCATACAGAGATA 38 76 361188 105 2123 CAGTAAGGTTTTCATACAGA 22 77 361189 105 2127 GAAGCAGTAAGGTTTTCATA 46 78 180307 105 2131 GAGAGAAGCAGTAAGGTTTT 32 79 361190 105 2212 GCTTTTCCTAGCTCTTTGAT 74 80 361191 105 2215 ATGGCTTTTCCTAGCTCTTT 68 81 361192 105 2347 ATGGTCTTATCCAAAAATGT 63 82 361193 105 2351 ACTCATGGTCTTATCCAAAA 66 83 361194 105 2355 CAATACTCATGGTCTTATCC 54 84 361195 105 2359 AATTCAATACTCATGGTCTT 69 85 361196 105 2383 ATGATTTCAGCTAACATCTC  1 86 180311 105 2386 GTGATGATTTCAGCTAACAT 59 87 361197 105 2407 GAATATTTTGGTATCTGATT 59 88 361198 105 2411 ATTTGAATATTTTGGTATCT 20 89 361199 105 2415 TTCCATTTGAATATTTTGGT 65 90 361200 105 2419 ATATTTCCATTTGAATATTT 51 91 361202 105 2425 TTTTTGATATTTCCATTTGA 20 92

Gap-widened oligonucleotides having the same sequences as the compounds described in Table 4 were also tested. All compounds in Table 4 are chimeric oligonucleotides (“gapmers”) 20 nucleotides in length, composed of a central “gap” region consisting of 16 2′-deoxynucleotides, which is flanked on both sides (5′ and 3′) by two-nucleotide “wings”. The wings are composed of 2′-O-(2-methoxyethyl)nucleotides, also known as 2′-MOE nucleotides. The internucleoside (backbone) linkages are phosphorothioate throughout the oligonucleotide. All cytidine residues are 5-methylcytidines. Shown in Table 4 is the sequence of the oligonucleotide, and the target site which is the first (5′ most) position on the target sequence to which the compound binds. The 2-16-2 motif compounds were analyzed for their effect on gene target mRNA levels by quantitative real-time PCR as described herein.

Data are averages from three experiments in which HepG2 cells were treated with 50 nM of the disclosed oligomeric compounds using LIPOFECTIN™. A reduction in expression is expressed as percent inhibition in Table 4. If present, “N.D.” indicates “not determined”. The target regions to which these oligomeric compounds are inhibitory are herein referred to as “validated target segments.”

TABLE 4 Inhibition of human GCCR mRNA levels by 2-16-2 gapmers ISIS Target % SEQ No of SEQ ID Target Inhib ID 2-16-2 NO Site Sequence w/2-16-2 NO 372350 105  394 TCTGTCTCTCCCATATACAG 69  5 372376 105  398 TGTTTCTGTCTCTCCCATAT 72  6 372331 105  402 CTTTTGTTTCTGTCTCTCCC 67  7 372341 105  406 ATCACTTTTGTTTCTGTCTC 63  8 352983 105  497 GTTTGCAATGCTTTCTTCCA 64  9 372365 105  501 TGAGGTTTGCAATGCTTTCT 69 10 372387 105  505 CTATTGAGGTTTGCAATGCT 70 11 372316 105  509 CGACCTATTGAGGTTTGCAA 73 12 372310 105  514 CTGGTCGACCTATTGAGGTT 70 13 372315 105  672 CTGTGGTATACAATTTCACA 35 14 372326 105  679 CTTTGGTCTGTGGTATACAA 54 15 372339 105  689 GTCAAAGGTGCTTTGGTCTG 81 16 372322 105  877 GGTTTAGTGTCCGGTAAAAT 78 17 372361 105  954 CTTTTTCTGTTTTCACTTGG 70 18 372308 105 1000 TTCTCTTGCTTAATTACCCC 84 19 372304 105 1004 CAGTTTCTCTTGCTTAATTA 66 20 352984 105 1007 GCCCAGTTTCTCTTGCTTAA 80 21 372372 105 1058 TTTATTACCAATTATATTTG 0 22 372327 105 1062 ACATTTTATTACCAATTATA 11 23 372311 105 1066 GCAGACATTTTATTACCAAT 65 24 372352 105 1070 AATGGCAGACATTTTATTAC 54 25 372337 105 1074 CAGAAATGGCAGACATTTTA 36 26 372323 105 1078 TGAACAGAAATGGCAGACAT 73 27 372347 105 1081 CCATGAACAGAAATGGCAGA 86 28 372383 105 1085 CACACCATGAACAGAAATGG 73 29 372348 105 1089 TACTCACACCATGAACAGAA 82 30 372363 105 1093 GAGGTACTCACACCATGAAC 47 31 372334 105 1097 TCCAGAGGTACTCACACCAT 82 32 372359 105 1101 GTCCTCCAGAGGTACTCACA 69 33 372344 105 1105 ATCTGTCCTCCAGAGGTACT 72 34 372307 105 1109 GTACATCTGTCCTCCAGAGG 74 35 372370 105 1113 AGTGGTACATCTGTCCTCCA 69 36 372374 105 1117 TCATAGTGGTACATCTGTCC 0 37 372355 105 1121 CATGTCATAGTGGTACATCT 65 38 372385 105 1125 TATTCATGTCATAGTGGTAC 18 39 372319 105 1129 GCTGTATTCATGTCATAGTG 23 40 372366 105 1133 GGATGCTGTATTCATGTCAT 37 41 372330 105 1137 AAAGGGATGCTGTATTCATG 80 42 372333 105 1141 TGAGAAAGGGATGCTGTATT 68 43 372358 105 1181 TGGTGGAATGACATTAAAAA 67 44 372381 105 1185 GAATTGGTGGAATGACATTA 30 45 372377 105 1324 GAGCTTACATCTGGTCTCAT 45 46 372309 105 1328 AGGAGAGCTTACATCTGGTC 63 47 372388 105 1332 ATGGAGGAGAGCTTACATCT 55 48 372321 105 1336 CTGGATGGAGGAGAGCTTAC 51 49 372312 105 1339 GAGCTGGATGGAGGAGAGCT 60 50 372324 105 1468 TGTCCTTCCACTGCTCTTTT 73 51 372332 105 1472 GTGCTGTCCTTCCACTGCTC 81 52 372335 105 1476 AATTGTGCTGTCCTTCCACT 42 53 372342 105 1480 AGGTAATTGTGCTGTCCTTC 100 54 372345 105 1543 CGGCATGCTGGGCAGTTTTT 82 55 372356 105 1547 ATAGCGGCATGCTGGGCAGT 73 56 372305 105 1549 CGATAGCGGCATGCTGGGCA 80 57 372367 105 1570 ATTCCAGCCTGAAGACATTT 78 58 372353 105 1574 GTTCATTCCAGCCTGAAGAC 70 59 372364 105 1597 TTCTTTGTTTTTCGAGCTTC 47 60 372340 105 1601 TTTTTTCTTTGTTTTTCGAG 100 61 372369 105 1680 CAGGAACTATTGITTTGTTA 56 62 372378 105 1682 TGCAGGAACTATTGTTTTGT 41 63 372317 105 1765 GAGCTATCATATCCTGCATA 84 64 372351 105 1769 AACAGAGCTATCATATCCTG 69 65 372389 105 1773 CTGGAACAGAGCTATCATAT 76 66 372362 105 1840 TTCACTGCTGCAATCACTTG 64 67 372328 105 1844 CCATTTCACTGCTGCAATCA 81 68 372338 105 1848 TTGCCCATTTCACTGCTGCA 82 69 372349 105 1999 ATAATCAGATCAGGAGCAAA 10 70 372373 105 2003 ATTAATAATCAGATCAGGAG 30 71 372360 105 2007 GCTCATTAATAATCAGATCA 27 72 372384 105 2011 CTCTGCTCATTAATAATCAG 100 73 372380 105 2015 CATTCTCTGCTCATTAATAA 2 74 372320 105 2053 AGCATGTGTTTACATTGGTC 75 75 372371 105 2119 AAGGTTTTCATACAGAGATA 37 76 372382 105 2123 CAGTAAGGTTTTCATACAGA 44 77 372306 105 2127 GAAGCAGTAAGGTTTTCATA 48 78 372343 105 2131 GAGAGAAGCAGTAAGGTTTT 46 79 372313 105 2212 GCTTTTCCTAGCTCTTTGAT 66 80 372325 105 2215 ATGGCTTTTCCTAGCTCTTT 69 81 372336 105 2347 ATGGTCTTATCCAAAAATGT 65 82 372318 105 2351 ACTCATGGTCTTATCCAAAA 70 83 372375 105 2355 CAATACTCATGGTCTTATCC 85 84 372346 105 2359 AATTCAATACTCATGGTCTT 47 85 372386 105 2383 ATGATTTCAGCTAACATCTC 74 86 372354 105 2386 GTGATGATTTCAGCTAACAT 66 87 372357 105 2407 GAATATTTTGGTATCTGATT 13 88 372368 105 2411 ATTTGAATATTTTGGTATCT 0 89 372379 105 2415 TTCCATTTGAATATTTTGGT 44 90 372390 105 2419 ATATTTCCATTTGAATATTT 0 91 372329 105 2425 TTTTTGATATTTCCATTTGA 0 92

The 2-16-2 oligonucleotides shown in Table 4 and the 5-10-5 oligonucleotides shown in Table 3 which reduced GCCR expression by at least 30% are preferred. The target segments to which these preferred sequences are complementary are herein referred to as “preferred target segments” and are therefore preferred for targeting by compounds of the present invention.

Example 7 Cross-Species Oligonucleotides Targeting GCCR

Some oligonucleotides described in the previous example are complementary across species and are therefore expected to reduce expression of glucocorticoid receptor across species. Shown in Table 5 is the sequence of such cross-species oligonucleotides, and the ISIS numbers of the 5-10-5 motif version and the 2-16-2 motif version of the oligonucleotide. Also indicated for each sequence is the target site which is the first (5′ most) position on the human target sequence (NM000176.1, incorporated herein as SEQ ID NO: 105) to which the compound binds. The complementarity for human, cynomolgus monkey, rat, and mouse GCCR mRNA is indicated (“yes” means perfect complementarity and “1 mm” means one mismatch from perfect complementarity).

TABLE 5 Cross-species oligonucleotides targeted to GCCR Pos'n ISIS # of ISIS # of SEQ on 5-10-5 2-16-2 ID SEQ ID Perfect complement to: gapmer gapmer NO Sequence NO: 1 Human Monkey Rat Mouse 361137 372316 12 cgacctattgaggtttgcaa  509 yes yes yes yes 180276 372326 15 ctttggtctgtggtatacaa  679 yes 1 mm 1 mm yes 345198 372339 16 gtcaaaggtgctttggtctg  689 yes yes yes yes 180304 372320 75 agcatgtgtttacattggtc 2053 yes yes yes yes 180275 372315 14 ctgtggtatacaatttcaca  672 yes 1 mm 1 mm yes 361141 372311 24 gcagacattttattaccaat 1066 yes yes yes 1 mm 180281 352984 21 gcccagtttctcttgcttaa 1007 yes yes yes yes 361151 372307 35 gtacatctgtcctccagagg 1109 yes yes yes yes 180274 372310 13 ctggtcgacctattgaggtt  514 yes yes yes yes 361156 372319 40 gctgtattcatgtcatagtg 1129 yes yes yes yes

Example 8 Antisense Inhibition of Human and Rat GCCR mRNA Levels—Dose-Response Studies with 5-10-5 Gapmers

In a further embodiment of the present invention, eleven oligonucleotides were selected for additional dose-response studies. Primary rat hepatocytes were treated with 5, 10, 25, 50, 100 or 200 nM of ISIS 180274, ISIS 180275, ISIS 180276, ISIS 180281, ISIS 180304, ISIS 361137, ISIS 361141, ISIS 361151, ISIS 361156, ISIS 345198, ISIS 361137 or the negative control oligonucleotide ISIS 141923 (CCTTCCCTGAAGGTTCCTCC, incorporated herein as SEQ ID NO: 2), and mRNA levels were measured as described in other examples herein. ISIS 141923 is a 5-10-5 gapmer comprising a ten deoxynucleotide gap flanked by 2′-MOE wings and a phosphorothioate backbone. All cytosines are 5-methylcytosines. Untreated cells served as the control to which the data were normalized.

Results of these studies are shown in Table 6. Target mRNA levels were measured by real-time PCR as described herein. Data are averages from three experiments and are expressed as percent inhibition relative to untreated control.

TABLE 6 Dose-dependent inhibition of GCCR expression in rat primary hepatocytes % Inhibition Dose of Oligonucleotide (nM) ISIS # SEQ ID NO 5 10 25 50 100 200 180274 13 16 33 29 65 84 89 180275 14 0 13 56 84 84 90 180276 15 23 43 43 68 89 93 180281 21 0 20 33 75 86 87 180304 75 42 51 47 75 86 91 361137 12 40 30 48 81 83 89 361141 24 36 61 48 77 87 92 361151 35 10 28 42 77 90 94 361156 40 22 47 46 66 84 92 345198 16 0 35 53 81 77 85 361158 42 34 50 47 79 91 93 141923 2 0 10 18 43 0 12

In a further embodiment of the present invention, the same oligonucleotides were tested in the human HepG2 cell line for their ability to reduce GCCR mRNA expression at the indicated doses. Untreated cells served as the control to which the data were normalized.

Results of these studies are shown in Table 7. Target mRNA levels were measured by real-time PCR as described herein. Data are averages from three experiments and are expressed as percent inhibition relative to untreated control.

TABLE 7 Dose-dependent inhibition of GCCR expression in HepG2 cells % Inhibition Dose of Oligonucleotide (nM) ISIS # SEQ ID NO 1 10 25 50 100 200 180274 13 0 31 54 66 77 83 180275 14 13 54 75 86 93 94 180276 15 26 77 87 92 94 98 180281 21 3 46 68 80 90 84 180304 75 0 64 90 90 92 91 361137 12 18 71 84 91 92 86 361141 24 1 49 81 85 73 78 361151 35 22 42 71 82 89 91 361156 40 7 75 75 79 80 82 345198 16 17 71 79 86 80 82 361158 42 11 35 78 80 82 77 141923 2 15 12 20 12 14 3

As shown in Table 6 and Table 7, antisense oligonucleotides targeting GCCR are effective at reducing both human and rat target mRNA levels in a dose-dependent manner in vitro.

Example 9 Antisense Inhibition of Rat GCCR mRNA Levels—In Vivo Dose-Response Studies with 5-10-5 Gapmers

Five of the 5-10-5 gapmer motif oligonucleotides (ISIS 180281, ISIS 361137, ISIS 345198, ISIS 180304, and ISIS 361141) were evaluated at various doses in rats for their ability to reduce GCCR mRNA levels in liver. Eight week-old Sprague Dawley rats were divided into treatment groups which received doses of 50, 25 or 12.5 mg/kg of one the indicated oligonucleotides via injection. Each treatment group was comprised of four animals, and was dosed twice weekly for 3 weeks. Animals injected with saline alone served as a control group. The animals were evaluated weekly for standard blood parameters (ALT/AST, cholesterol, triglycerides, and glucose). Animals were sacrificed at the end of the study and liver tissue was collected and analyzed for target reduction using real-time PCR analysis methods described herein. Results are shown in Tables 8a and 8b (separate experiments) as the percentage reduction in GCCR mRNA measured after treatment with the indicated doses of the indicated oligonucleotides.

TABLE 8a In vivo rat screen- GCCR antisense oligonucleotides % Reduction in GCCR mRNA in rat liver (compared to saline-treated controls) Compound 50 mg/kg 25 mg/kg 12.5 mg/kg ISIS 180281 68 65 48 ISIS 180304 52 34 0 ISIS 345198 63 58 52

TABLE 8b In vivo rat screen- GCCR antisense oligonucleotides % Reduction in GCCR mRNA in rat liver (compared to saline-treated controls) Compound 50 mg/kg 25 mg/kg 12.5 mg/kg ISIS 180281 62 62 59 ISIS 361137 59 47 32 ISIS 361141 61 49 22

The data in Tables 8a and 8b show that antisense oligonucleotides targeted to GCCR are effective at reducing expression in vivo in a dose-dependent manner. ISIS 345198 (GTCAAAGGTGCTTTGGTCTG; SEQ ID NO: 16) was chosen for further evaluation in structure-activity experiments focusing on gap optimization. This compound is perfectly complementary to mouse, rat, human, monkey, rabbit and guinea pig glucocorticoid receptor RNA.

Example 10 Antisense Inhibition of GCCR mRNA Levels In Vivo—Gap Optimization Study

A series of oligomeric compounds were designed to target GCCR with varying sizes of the deoxynucleotide gap and 2′-MOE wings. Each of the oligonucleotides tested has the same nucleobase sequence (GTCAAAGGTGCTTTGGTCTG, incorporated herein as SEQ ID NO: 16) and therefore targets the same segment of SEQ ID NO: 105 (nucleobases 689 to 709). As shown in Example 7, this oligonucleotide is also perfectly complementary to rat GCCR.

The compounds are shown in Table 9. Plain text indicates a deoxynucleotide, and nucleobases designated with bold, underlined text are 2′-O-(2-methoxyethyl)nucleotides. Internucleoside linkages are phosphorothioate throughout, and all cytosines are 5-methylcytosines.

Indicated in Table 9 is the “motif' of each compound indicative of chemically distinct regions comprising the oligonucleotide.

TABLE 9 Antisense compounds targeting rat GCCR ISIS Number Chemistry Motif 345198 GTCAAAGGTGCTTTGGTCTG 5-10-5 gapmer 372339 GTCAAAGGTGCTTTGGTCTG 2-16-2 gapmer 377130 GTCAAAGGTGCTTTGGTCTG 3-14-3 gapmer 377131 GTCAAAGGTGCTTTGGTCTG 4-12-4 gapmer

Nine-week old Sprague-Dawley male rats were treated twice weekly for three weeks with doses of 50, 25, 12.5, and 6.25 mg/kg of the oligonucleotides presented in Table 9. Animals injected with saline alone served as controls. Each treatment group was comprised of four animals.

At the end of the study, animals were sacrificed, and tissues were collected for determination of target reduction and oligonucleotide concentration.

White adipose tissue was analyzed for target reduction using real-time PCR analysis methods described herein. Results are shown in Tables 10a, 10b, and 10c (separate experiments) as the percentage reduction in GCCR mRNA measured after treatment with the indicated doses of the indicated oligonucleotides. Tissues from animals treated with each gap-widened oligonucleotide were assayed for target reduction alongside tissues from animals treated with the 5-10-5 motif oligonucleotide for comparison.

TABLE 10a In vivo reduction of GCCR levels in white adipose tissue with 2-16-2 oligonucleotides % Inhibition Treatment Dose of oligonucleotide (mg/kg) group 50 25 12.5 6.25 ISIS 345198 56 26 17 7 ISIS 372339 34 0 8 8

TABLE 10b In vivo reduction of GCCR levels in white adipose tissue with 3-14-3 oligonucleotides % Inhibition Treatment Dose of oligonucleotide (mg/kg) group 50 25 12.5 6.25 ISIS 345198 59 49 27 22 ISIS 377130 54 37 21 18

TABLE 10c In vivo reduction of GCCR levels in white adipose tissue with 4-12-4 oligonucleotides % Inhibition Treatment Dose of oligonucleotide (mg/kg) group 50 25 12.5 6.25 ISIS 345198 56 23 21 7 ISIS 377131 55 23 15 0

Liver tissue was also analyzed for target reduction using real-time PCR analysis methods described herein. Results are shown in Tables 11a, 11b, and 11c (separate experiments) as the percentage reduction in GCCR mRNA measured after treatment with the indicated doses of the indicated oligonucleotides. Tissues from animals treated with each gap-widened oligonucleotide were assayed for target reduction alongside tissues from animals treated with the 5-10-5 motif oligonucleotide for comparison.

TABLE 11a In vivo reduction of GCCR levels in liver with 2-16-2 oligonucleotides % Inhibition Treatment Dose of oligonucleotide (mg/kg) group 50 25 12.5 6.25 ISIS 345198 78 77 65 51 ISIS 372339 83 77 56 44

TABLE 11b In vivo reduction of GCCR levels in liver with 3-14-3 oligonucleotides % Inhibition Treatment Dose of oligonucleotide (mg/kg) group 50 25 12.5 6.25 ISIS 345198 78 80 67 54 ISIS 377130 87 78 68 43

TABLE 11c In vivo reduction of GCCR levels in liver with 4-12-4 oligonucleotides % Inhibition Treatment Dose of oligonucleotide (mg/kg) group 50 25 12.5 6.25 ISIS 345198 76 75 58 49 ISIS 377131 82 64 60 61

As shown in Tables 11a, 11b, and 11c, all of the gap-widened oligonucleotides tested were effective at reducing GCCR levels in a dose-dependent manner in vivo. In addition, the gap-widened oligonucleotides show a trend toward greater potency than the 5-10-5 gapmer in the liver.

In addition, to determine effects of altering the gap size on pharmacokinetics, oligonucleotide concentration in kidney and liver were determined. Methods to determine oligonucleotide concentration in tissues are known in the art (Geary et al., Anal Biochem, 1999, 274, 241-248). Total oligonucleotide is the sum of all oligonucleotides metabolites detected in the tissue. Shown in Table 12 are the total concentration and the concentration of full length oligonucleotide (in μg/g) in the liver of animals treated with the indicated oligonucleotide at the indicated concentration.

TABLE 12 GCCR oligonucleotide concentration in rat liver Liver Liver Total Full- Treatment Motif Dose oligo length ISIS 345198 5-10-5 25 mg/kg 507 408 12.5 mg/kg 318 224 ISIS 372339 2-16-2 25 mg/kg 450 306 12.5 mg/kg 311 183 ISIS 377130 3-14-3 25 mg/kg 575 315 12.5 mg/kg 350 212 ISIS 377131 4-12-4 25 mg/kg 584 424 12.5 mg/kg 354 265

As shown in Table 12, the levels of full-length oligonucleotide in the liver are comparable or reduced for ISIS 372339 and ISIS 377130 as compared to ISIS 345198. Coupled with the target reduction as shown in Table 11, these data show that the enhanced potency of the gap-widened compounds is not due to enhanced accumulation of the compound in the liver. Thus, preferred oligonucleotides of the present invention include gap-widened oligonucleotides that show enhanced or comparable potency with regard to target reduction to the corresponding 5-10-5 gapmer without enhanced accumulation of the compound in a target tissue. In some embodiments, the target tissue is adipose and in some embodiments, the target tissue is liver.

Example 11 Design of “Gap-Widened” Antisense Oligonucleotides Targeting Human GCGR

A series of oligomeric compounds were designed to target human GCGR (Genbank accession number: NM000160.1, incorporated herein as SEQ ID NO: 108), with varying sizes of the deoxynucleotide gap and 2′-MOE wings. Each of the oligonucleotides is 20 nucleobases in length and has the same nucleobase sequence (GCACTTTGTGGTGCCAAGGC, incorporated herein as SEQ ID NO: 93), and therefore targets the same segment of SEQ ID NO: 108 (nucleobases 532 to 551). The compounds are shown in Table 13. Plain text indicates a deoxynucleotide, and nucleotides designated with bold, underlined text are 2′-O-(2-methoxyethyl)nucleotides. Internucleoside linkages are phosphorothioate throughout, and all cytosines are 5-methylcytosines. Indicated in Table 13 is the “motif' of each compound, indicative of chemically distinct regions comprising the oligonucleotide.

TABLE 13 Antisense compounds targeting human GCGR ISIS Number Chemistry Motif 310457 GCACTTTGTGGTGCCAAGGC 5-10-5 gapmer 325448 GCACTTTGTGGTGCCAAGGC 2-16-2 gapmer 325568 GCACTTTGTGGTGCCAAGGC 3-14-3 gapmer

The 5-10-5 gapmer, ISIS 310457, was tested for its ability to reduce target mRNA levels in vitro. HepG2 cells were treated with ISIS 310457 using methods as described herein. ISIS 310457 was analyzed for its effect on human glucagon receptor mRNA levels by quantitative real-time PCR and was found to reduce expression of GCGR by about 96%.

Example 12 Design of “Gap-Widened” Antisense Oligonucleotides Targeting Rat GCGR

A series of oligomeric compounds were designed to target rat GCGR (Genbank accession number: M96674.1, incorporated herein as SEQ ID NO: 109) with varying sizes of the deoxynucleotide gap and 2′-MOE wings. Each of the oligonucleotides tested has the same nucleobase sequence (GCACTTTGTGGTACCAAGGT, incorporated herein as SEQ ID NO: 94) and therefore targets the same segment of SEQ ID NO: 109 (nucleobases 402 to 421). The segment targeted by the rat oligonucleotides corresponds to the segment of human GCGR targeted by ISIS 310457 (SEQ ID NO: 93). The compounds are shown in Table 14. Plain text indicates a deoxynucleotide, and nucleotides designated with bold, underlined text are 2′-O-(2-methoxyethyl)nucleotides. Internucleoside linkages are phosphorothioate throughout, and all cytosines are 5-methylcytosines. Indicated in Table 14 is the “motif' of each compound indicative of chemically distinct regions comprising the oligonucleotide.

TABLE 14 Antisense compounds targeting rat GCGR ISIS Number Chemistry Motif 356171 GCACTTTGTGGTACCAAGGT 5-10-5 gapmer 357368 GCACTTTGTGGTACCAAGGT Uniform deoxy 357369 GCACTTTGTGGTACCAAGGT 1-18-1 gapmer 357370 GCACTTTGTGGTACCAAGGT 1-17-2 gapmer 357371 GCACTTTGTGGTACCAAGGT 2-16-2 gapmer 357372 GCACTTTGTGGTACCAAGGT 3-14-3 gapmer 357373 GCACTTTGTGGTACCAAGGT 4-12-4 gapmer

Example 13 Effects of Antisense Oligonucleotides Targeting GCGR—In Vivo Rat Study

In accordance with the present invention, the oligonucleotides designed to target rat GCGR were tested in vivo. Male Sprague Dawley rats, eight weeks of age, were injected with 50, 25, 12.5, or 6.25 mg/kg of ISIS 356171, ISIS 357368, ISIS 357369, ISIS 357370, ISIS 357371, ISIS 357372, or ISIS 357373 twice weekly for 3 weeks for a total of 6 doses. Saline-injected animals served as a control. Each of the oligonucleotides tested has the same nucleobase sequence (GCACTTTGTGGTACCAAGGT, incorporated herein as SEQ ID NO: 94), and the chemistry and motif of each compound is described above.

After the treatment period, rats were sacrificed and target nucleic acid levels were evaluated in liver. RNA isolation and target mRNA expression level quantitation are performed as described by other examples herein using RIBOGREEN™. RNA from each treatment group was assayed alongside RNA from the group treated with ISIS 356171. Results are presented in Table 15a, 15b, 15c, 15d, 15e, and 15f as a percentage of saline-treated control levels.

TABLE 15a Reduction of target levels in liver of rats treated with 2-16-2 antisense oligonucleotides targeted to GCGR % Control Dose of oligonucleotide (mg/kg) Treatment Motif 50 25 12.5 6.25 ISIS 356171 5-10-5 7 20 26 36 ISIS 357371 2-16-2 11 22 35 39

TABLE 15b Reduction of target levels in liver of rats treated with 3-14-3 antisense oligonucleotides targeted to GCGR % Control Dose of oligonucleotide (mg/kg) Treatment Motif 50 25 12.5 6.25 ISIS 356171 5-10-5 10 24 28 50 ISIS 357372 3-14-3 12 23 37 56

TABLE 15c Reduction of target levels in liver of rats treated with 4-12-4 antisense oligonucleotides targeted to GCGR % Control Dose of oligonucleotide (mg/kg) Treatment Motif 50 25 12.5 6.25 ISIS 356171 5-10-5 10 25 36 47 ISIS 357373 4-12-4 13 22 48 47

TABLE 15d Reduction of target levels in liver of rats treated with 1-17-2 antisense oligonucleotides targeted to GCGR % Control Dose of oligonucleotide (mg/kg) Treatment Motif 50 25 12.5 6.25 ISIS 356171 5-10-5 8 24 32 43 ISIS 357370 1-17-2 20 41 62 68

TABLE 15e Reduction of target levels in liver of rats treated with 1-18-1 antisense oligonucleotides targeted to GCGR % Control Dose of oligonucleotide (mg/kg) Treatment Motif 50 25 12.5 6.25 ISIS 356171 5-10-5 9 27 34 46 ISIS 357369 1-18-1 33 35 58 70

TABLE 15f Reduction of target levels in liver of rats treated with uniform deoxy oligonucleotides targeted to GCGR % Control Dose of oligonucleotide (mg/kg) Treatment Motif 50 25 12.5 6.25 ISIS 356171 5-10-5 8 23 30 45 ISIS 357368 Uniform deoxy 31 43 77 73

As shown in Tables 15a, 15b, 15c, 15d, and 15e the gap-widened antisense oligonucleotides were effective at reducing GCGR levels in vivo in a dose-dependent manner.

In addition, oligonucleotide concentration in kidney and liver were determined. Methods to determine oligonucleotide concentration in tissues are known in the art (Geary et al., Anal. Biochem., 1999, 274, 241-248). Shown in Table 16 are the total oligonucleotide concentration and the concentration of full length oligonucleotide (in μg/g) in the kidney or liver of animals treated with 25 mg/kg of the indicated oligonucleotide. Total oligonucleotide is the sum of all oligonucleotides metabolites detected in the tissue.

TABLE 16 Concentration of oligonucleotide in liver and kidney Kidney Kidney Liver Liver Total Full- Total Full- Treatment Motif oligo length oligo length ISIS 356171 5-10-5 gapmer 1814 1510 621 571 ISIS 356368 Uniform deoxy 801 183 282 62 ISIS 356369 1-18-1 1237 475 309 171 ISIS 356370 1-17-2 1127 590 370 271 ISIS 356371 2-16-2 871 515 345 253 ISIS 356372 3-14-3 1149 774 497 417 ISIS 356373 4-12-4 902 687 377 326

As shown in Table 16, the concentrations of the gap-widened oligonucleotides in kidney were generally reduced with respect to those found for ISIS 356171 in these tissues. Taken with the target reduction data shown in Table 15 wherein potency was maintained with ISIS 356371, ISIS 356372, and ISIS 356373 with respect to ISIS 356171, these data suggest that gap-widened oligos, particularly ISIS 356371, ISIS 356372, and ISIS 356373 are, in essence, more effective than ISIS 356171 at reducing target levels in the liver.

Example 14 Effects of Antisense Oligonucleotides Targeting GCGR—In Vivo Study in Cynomolgus Monkeys

To evaluate alterations in tissue distribution, potency, or therapeutic index caused by modification of the antisense oligonucleotide motif in a primate, cynomolgus monkeys were injected with ISIS 310457 (5-10-5 motif) or ISIS 325568 (2-16-2 motif) at doses of 3, 10, or 20 mg/kg per week. These antisense compounds show 100% complementarity to the monkey GCGR target sequence. Animals injected with saline alone served as controls. The duration of the study was 7 weeks, and the animals were dosed three times during the first week, followed by once-weekly dosing for 6 weeks. Each treatment group was comprised of 5 animals. One group treated with 20 mg/kg of ISIS 310457 and one group treated with 20 mg/kg of ISIS 325568 recovered for three weeks after cessation of dosing prior to sacrifice (”20 mg/kg recovery“). Other treatment groups were sacrificed at the end of the study. Liver tissues were collected to assess target reduction.

RNA isolation and target mRNA expression level quantitation were performed as described by other examples herein using RIBOGREEN™. Results are presented in Table 17 as a percentage of saline-treated control levels.

TABLE 17 Reduction of target levels in liver of monkeys treated with antisense oligonucleotides targeted to GCGR % Control Dose of oligonucleotide 20 mg/kg, Treatment Motif recovery 20 mg/kg 10 mg/kg 3 mg/kg ISIS 310457 5-10-5 27 34 43 71 ISIS 325568 2-16-2 43 45 54 49

As shown in Table 17, treatment with ISIS 310457 and 325568 caused decreases in GCGR levels at all of the doses tested, and reduction in target levels was still observed in the 20 mg/kg recovery groups. ISIS 325568 caused greater reduction than ISIS 310457 at the 3 mg/kg dose.

In addition, oligonucleotide concentration in kidney and liver were determined. Methods to determine oligonucleotide concentration in tissues are known in the art (Geary et al., Anal Biochem, 1999, 274, 241-248). Shown in Table 18 are the total concentration and the concentration of full length oligonucleotide (in μg/g) in the kidney or liver of animals treated with the indicated oligonucleotide.

TABLE 18 Concentration of oligonucleotide in liver and kidney Kidney Kidney Liver Liver Total Full- Total Full- Treatment Motif Dose oligo length oligo length ISIS 310457 5-10-5  3 mg/kg 471 423 449 330 10 mg/kg 1011 911 710 606 20 mg/kg 1582 1422 981 867 20 mg/kg 449 347 648 498 recovery ISIS 325568 2-16-2  3 mg/kg 356 298 309 228 10 mg/kg 830 685 477 339 20 mg/kg 1390 1101 739 544 20 mg/kg 264 161 344 205 recovery

As shown in Table 18, the kidney concentration of the 5-10-5 motif oligonucleotide ISIS 310457 is higher than that measured for the 2-16-2 motif oligonucleotide ISIS 325568 at all concentrations tested. Taken with the target reduction data in Table 9 for the 2-16-2 motif oligonucleotide, these data suggest that the gap-widened oligonucleotide is more potent than the corresponding 5-10-5 motif oligonucleotide, providing a more robust lowering of target mRNA levels in the liver without enhanced accumulation of oligonucleotide.

Example 15 Effects of Gap-Widened Oligonucleotides on Reduction of DGAT2 mRNA Levels—In Vitro Analysis

In accord with the present invention, oligonucleotides were designed to target DGAT2.

Shown in Table 19 is the sequence of each oligonucleotide. Plain text indicates a deoxynucleotide, and nucleotides designated with bold, underlined text are 2′-O-(2-methoxyethyl)nucleotides. Also shown for each oligonucleotide in Table 19 is its motif, the target site on human DGAT2 mRNA (GENBANK® accession number NM032564.2, incorporated herein as SEQ ID NO: 110), and its cross-species identity. For each species listed, an “X” denotes perfect complementarity to the target sequence for that species, “1 MM” denotes one mismatch to the target sequence for the species, etc.

TABLE 19 Antisense compounds targeting DGAT2 SEQ ID Target ISIS # Sequence NO Site Motif Human Monkey Rat Mouse 217328 GCATTGCCACTCCCATTCTT  95  909 5-10-5 X X  1 MM X 334177 AGGACCCCGGAGTAGGCGGC  96  246 5-10-5 X X  1 MM X 366710 GACCTATTGAGCCAGGTGAC  97  396 5-10-5 X X  2 MM  2 MM 366714 GTAGCTGCTTTTCCACCTTG  98  416 5-10-5 X X  2 MM  3 MM 370727 AGCTGCTTTTCCACCTTGGA  99  414 2-16-2 X X  2 MM  2 MM 370747 TGGAGCTCAGAGACTCAGCC 100  953 2-16-2 X X  3 MM  2 MM 370784 GCTGCATCCATGTCATCAGC 101 2099 2-16-2 X X >3 MM >3 MM

Each of these oligonucleotides was tested in vitro for their ability to reduce human DGAT2 mRNA levels using real time RT-PCR methods as described herein. In HepG2 and A549 cells, each of the oligonucleotides in Table 19 demonstrated IC50 values of about 20 nM.

Example 16 Effects of Gap-Widened Oligonucleotides on Reduction of DGAT2 mRNA Levels—In Vivo Analysis

The oligonucleotides described in Table 19, along with ISIS 217357 (ACACACTAGAAGTGAGCTTA, SEQ ID NO: 102), which is targeted to rat DGAT2, the complement of nucleotides 15333000 to 15365000 of GENBANK® accession number NW047561.1, herein incorporated as SEQ ID NO: 111 were tested for their ability to reduce DGAT2 levels in vivo. Eight week-old male Sprague-Dawley rats were injected with 20 mg/kg of oligonucleotide per week for 2 weeks. Each treatment group was comprised of 6 animals. Animals injected with saline alone served as controls.

At the end of the treatment period, animals were sacrificed and liver and kidney tissues were harvested. To determine effects of altering the gap size on pharmacokinetics, oligonucleotide concentration in kidney and liver were determined. Methods to determine oligonucleotide concentration in tissues are known in the art (Geary et al., Anal Biochem, 1999, 274, 241-248). Total oligonucleotide is the sum of all oligonucleotides metabolites detected in the tissue. Shown in Table 20 are the total concentration and the concentration of full length oligonucleotide (in μg/g) in the liver of animals treated with the indicated oligonucleotide concentration.

TABLE 20 Concentration of DGAT2 oligonucleotides in rat liver and kidney Treatment Total Full length group Motif Liver Kidney Liver Kidney ISIS 217357 5-10-5 91 441 70 328 ISIS 217328 5-10-5 145 399 121 294 ISIS 334177 5-10-5 164 650 114 392 ISIS 366710 5-10-5 166 625 123 401 ISIS 366714 5-10-5 278 674 214 488 ISIS 370727 2-16-2 209 355 131 166 ISIS 370747 2-16-2 195 480 150 342 ISIS 370784 2-16-2 303 669 256 421

As shown in Table 20, kidney concentrations of gap-widened oligonucleotides, particularly ISIS 370727 and ISIS 370747, were generally lower than those of oligonucleotides with a 10-deoxynucleotide gap.

Example 17 Effects of Gap-Widened Oligonucleotides on Reduction of DGAT2 mRNA Levels—In Vivo Analysis

In another arm of the experiment described in Example 16, eight-week old male Sprague-Dawley rats were treated with the oligonucleotides at doses of 50 mg/kg per week for four weeks. Each treatment group was comprised of 4 animals. At the end of the treatment period, animals were sacrificed and target mRNA levels were determined using real-time RT-PCR as described herein. Results are shown in Table 21 as the average % inhibition for each treatment group.

TABLE 21 Reduction of target levels in rat liver with oligonucleotides targeting DGAT2 Treatment % group Motif Inhibition ISIS 217357 5-10-5 25 ISIS 217328 5-10-5 48 ISIS 334177 5-10-5 51 ISIS 366710 5-10-5 63 ISIS 366714 5-10-5 67 ISIS 370727 2-16-2 77 ISIS 370747 2-16-2 79 ISIS 370784 2-16-2 52

As shown in Table 21, the gap-widened oligonucleotides targeted to DGAT2 show excellent inhibitory activity in the liver. ISIS 370727 and ISIS 370747, in particular, showed superior ability to reduce target expression. Taken with the distribution of these oligonucleotides in the liver as shown in Table 20, these data suggest that gap-widened oligonucleotides provide excellent to superior target reduction without enhanced accumulation of oligonucleotide in target tissues. In addition, the gap-widened oligonucleotides possess a preferred liver to kidney ratio as compared to the 5-10-5 motif oligonucleotides targeting DGAT2.

Example 18 Effects of Gap-Widened Oligonucleotides on Reduction of CRP mRNA Levels—In Vivo Analysis

Monkey-human cross-species oligonucleotides targeted to C-reactive protein (CRP) were designed to target CRP using sequences known in the art (see US application publication number US2005-0014257, herein incorporated by reference in its entirety). Shown in Table 22 is the sequence of oligonucleotides targeted to CRP tested in cynomologus monkeys. Plain text indicates a deoxynucleotide, and nucleotides designated with bold, underlined text are 2′-O-(2-methoxyethyl)nucleotides. Also shown for each oligonucleotide in Table 22 is its motif.

TABLE 22 Antisense oligonucleotides targeting CRP SEQ ID Isis # Sequence NO Motif 353512 TCCCATTTCAGGAGACCTGG 115 3-14-3 330012 TCCCATTTCAGGAGACCTGG 115 5-10-5 353491 GCACTCTGGACCCAAACCAG 116 3-14-3 133726 GCACTCTGGACCCAAACCAG 116 5-10-5

Methods of assaying for activity of CRP compounds in vivo and in vitro are known in the art (see US application publication number US2005-0014257, herein incorporated by reference). Toxicity profiles of gap-widened oligonucleotides were compared to the 5-10-5 oligonucleotides by treating monkeys with 14 or 40 mg/kg/wk for 4 weeks. Activity was compared in a dose-escalation study with each cycle containing four subcutaneous doses administered (Mon., Wed., Fri., Mon.) in 4 dosing cycles over 8 weeks. Doses were 2, 4 and 10 mg/kg. At 48 hr following the last dose in each treatment cycle, monkeys were challenged with 1 to 2 μg/kg IL-6 (administered subcutaneously) and serum CRP levels were quantified over 36 hours. Serum CRP levels may be measured by ELISA using a commercially available kit (for example, ALerCHEK Inc., Portland, Me.). Animals were sacrificed after the second and fourth cycles and liver CRP mRNA, tissue oligonucleotide concentration, clinical signs, serum chemistry, hematology, body weight, and histology were assessed. With regard to tissue oligonucleotide concentration and histology, the primary difference was 30% lower kidney concentration and fewer histologic changes in the 3-14-3 treated animals. Plasma cytokine and CRP levels were examined but not significantly increased.

Several CRP inhibitors were pharmacologically active, with the greatest reductions in serum CRP (30-66%) and hepatic CRP mRNA (60-85%) observed at both the 4 and 10 mg/kg treatment cycles.

We have surprisingly found that chimeric antisense compounds with gaps at least 11 nucleobases long and wings which are from independently from 1 to 4 nucleobases in length which are 2′-MOE-modified. This enhanced efficacy is not predicted by the rank order potency of these compounds in vitro (cell culture). 2-16-2 and 3-14-3 gapmer compounds as well as 3-10-7 and 7-10-3 gapmer compounds have been shown to be more effective than 5-10-5 chimeras of the equivalent sequence and wing modification. 4-12-4 gapmers are also believed to be a useful embodiment.

DETAILED DESCRIPTION OF EMBODIMENTS

Non-limiting examples of 2′-modified nucleosides useful in the compounds of the present invention, include but are not limited to 2′-O-alkyl, 2′-O-alkyl-O-alkyl wherein alkyl is a C1 to C6 alkyl or C1 to C6 alkylene when alkyl is not a terminal substituent. These include 2′-O-methyl, 2′-O-propyl and 2′-O-methoxyethyl nucleosides.

Details

The present invention uses antisense compounds which are chimeric compounds. “Chimeric” antisense compounds or “chimeras,” in the context of this invention, are antisense compounds, particularly oligonucleotides, 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 oligonucleotide. These oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, increased stability and/or increased binding affinity for the target nucleic acid. An additional region of the oligonucleotide 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 oligonucleotide-mediated inhibition of gene expression. The cleavage of RNA:RNA hybrids can, in like fashion, be accomplished through the actions of endoribonucleases, such as RNAseL which cleaves both cellular and viral RNA. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.

Chimeric antisense compounds of the invention may be formed as compositestructures of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above. Such compounds have also been, referred to in the art as hybrids or gapmers. Representative United States patents that teach the preparation of such hybrid structures include, but are not limited to, U.S. Pat. Nos. 5,013,830; 5,149,797; 5,220,007; 5,256,775; 5,366,878; 5,403,711; 5,491,133; 5,565,350; 5,623,065; 5,652,355; 5,652,356; and 5,700,922, each of which is herein incorporated by reference in its entirety.

Synthesis of Nucleoside Phosphoramidites

The following compounds, including amidites and their intermediates were prepared as described in U.S. Pat. No. 6,426,220 and published PCT WO 02/36743; 5′-ODimethoxytrityl-thymidine intermediate for 5-methyl dC amidite, 5′-O-Dimethoxytrityl2′-deoxy-5-methylcytidine intermediate for 5-methyl-dC amidite, 5′-O-Dimethoxytrityl2′-deoxy-N4-benzoyl-5-methylcytidine penultimate intermediate for 5-methyl dC amidite, [5′-O-(4,4′-Dimethoxytriphenylmethyl)-2′-deoxy-N4-benzoyl-5-methylcytidin-3′O-yl]-2-cyanoethyl-N,N-diisopropylphosphoramidite(5-methyl dC amidite), 2′-Fluorodeoxyadenosine, 2′-Fluorodeoxyguanosine, 2′-Fluorouridine, 2′Fluorodeoxycytidine, 2′-O-(2-Methoxyethyl) modified amidites, 2′-O-(2-methoxyethyl)5-methyluridine intermediate, 5′-O-DMT-2′-O-(2-methoxyethyl)-5-methyluridine penultimate intermediate, [5′-O-(4,4′-Dimethoxytriphenylmethyl)-2′-O-(2-methoxyethyl)5-methyluridin-3′-O-yl]-2-cyanoethyl-N,N-diisopropylphosphoramidite (MOE T amidite), 5′-O-Dimethoxytrityl-2′-O-(2-methoxyethyl)-5-methylcytidine intermediate, 5′O-dimethoxytrityl-2′-O-(2-methoxyethyl)-N4-benzoyl-5-methyl-cytidine penultimate intermediate, [5′-O-(4,4′-Dimethoxytriphenylmethyl)-2′-O-(2-methoxyethyl)-N4-benzoyl5-methylcytidin-3′-O-yl]-2-cyanoethyl-N,N-diisopropylphosphoramidite (MOE 5-Me-C amidite), [5′-O-(4,4′-Dimethoxytriphenylmethyl)-2′-O-(2-methoxyethyl)-N6benzoyladenosin-3′-O-yl]-2-cyanoethyl-N,N-diisopropylphosphoramidite (MOE A amdite), [5′-O-(4,4′-Dimethoxytriphenylmethyl)-2′-O-(2-methoxyethyl)-N4isobutyrylguanosin-3′-O-yl]-2-cyanoethyl-N,N-diisopropylphosphoramidite (MOE G amidite), 2′-O-(Aminooxyethyl) nucleoside amidites and 2′-O-(dimethylaminooxyethyl)nucleoside amidites, 2′-(Dimethylaminooxyethoxy)nucleoside amidites, 5′-O-tertButyldiphenylsilyl-O2-2′-anhydro-5-methyluridine, 5′-O-tert-Butyldiphenylsilyl-2′-O-(2hydroxyethyl)-5-methyluridine, 2′-O-([2-phthalimidoxy)ethyl]-5′-t-butyldiphenylsilyl-5-methyluridine, 5′-O-tert-butyldiphenylsilyl-2′-O-[(2-formadoximinooxy)ethyl]-5methyluridine, 5′-O-tert-Butyldiphenylsilyl-2′-O-[N,N dimethylaminooxyethyl]-5methyluridine, 2′-O-(dimethylaminooxyethyl)-5-methyluridine, 5′-O-DMT-2′-O(dimethylaminooxyethyl)-5-methyluridine, 5′-O-DMT-2′-O-(2-N,Ndimethylaminooxyethyl)-5-methyluridine-3′-[(2-cyanoethyl)-N,N-diisopropylphosphoramidite], 2′-(Aminooxyethoxy)nucleoside amidites, N2-isobutyryl6-O-diphenylcarbamoyl-2′-O-(2-ethylacetyl)-5′-O-(4,4′-dimethoxytrityl)guanosine-3′-[(2cyanoethyl)-N,N-diisopropylphosphoramidite], 2′-dimethylaminoethoxyethoxy (2′DMAEOE) nucleoside amidites, 2′-O-[2(2-N,N-dimethylaminoethoxy)ethyl]-5-methyl uridine, 5′-O-dimethoxytrityl-2′-O-[2(2-N,N-dimethylaminoethoxy)-ethyl)]-5-methyl uridine and 5′-O-Dimethoxytrityl-2′-O-[2(2-N,N-dimethylaminoethoxy)-ethyl)]-5-methyl uridine-3′-O-(cyanoethyl-N,N-diisopropyl)phosphoramidite.

Oligonucleotide and Oligonucleoside Synthesis

The antisense compounds used in accordance with this invention may be conveniently and routinely made through the well-known technique of solid phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, Calif.). Any other means for such synthesis known in the art may additionally or alternatively be employed. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and alkylated derivatives. Oligonucleotides: Unsubstituted and substituted phosphodiester (P═O) oligonucleotides are synthesized on an automated DNA synthesizer (Applied Biosystems model 394) using standard phosphoramidite chemistry with oxidation by iodine. Phosphorothioates (P═S) are synthesized similar to phosphodiester oligonucleotides with the following exceptions: thiation was effected by utilizing a 10% w/v solution of 3,H-1,2-benzodithiole-3-one 1,1-dioxide in acetonitrile for the oxidation of the phosphite linkages. The thiation reaction step time was increased to 180 sec and preceded by the normal capping step. After cleavage from the CPG column and deblocking in concentrated ammonium hydroxide at 55° C. (12-16 hr), the oligonucleotides were recovered by precipitating with >3 volumes of ethanol from a 1 M NH4OAc solution. Phosphinate oligonucleotides are prepared as described in U.S. Pat. No. 5,508,270, herein incorporated by reference. Alkyl phosphonate oligonucleotides are prepared as described in U.S. Pat. No. 4,469,863, herein incorporated by reference.3′-Deoxy-3′-methylene phosphonate oligonucleotides are prepared as described in U.S. Pat. Nos. 5,610,289 or 5,625,050, herein incorporated by reference. Phosphoramidite oligonucleotides are prepared as described in U.S. Pat. No. 5,256,775 or U.S. Pat. No. 5,366,878, herein incorporated by reference. Alkylphosphonothioate oligonucleotides are prepared as described in published PCT applications PCT/US94/00902 and PCT/US93/06976 (published as WO 94/17093 and WO 94/02499, respectively), herein incorporated by reference. 3′-Deoxy-3′-amino phosphoramidate oligonucleotides are prepared as described in U.S. Pat. No. 5,476,925, herein incorporated by reference. Phosphotriester oligonucleotides are prepared as described in U.S. Pat. No. 5,023,243, herein incorporated by reference. Borano phosphate oligonucleotides are prepared as described in U.S. Pat. Nos. 5,130,302 and 5,177,198, both herein incorporated by reference. Oligonucleosides: Methylenemethylimino linked oligonucleosides, also identified as MMI linked oligonucleosides, methylenedimethylhydrazo linked oligonucleosides, also identified as MDH linked oligonucleosides, and methylenecarbonyl amino linked oligonucleosides, also identified as amide-3 linked oligonucleosides, and methyleneaminocarbonyl linked oligonucleosides, also identified as amide-4 linked oligonucleosides, as well as mixed backbone compounds having, for instance, alternating MMI and P═O or P═S linkages are prepared as described in U.S. Pat. Nos. 5,378,825, 5,386,023, 5,489,677, 5,602,240 and 5,610,289, all of which are herein incorporated by reference. Formacetal and thioformacetal linked oligonucleosides are prepared as described in U.S. Pat. Nos. 5,264,562 and 5,264,564, herein incorporated by reference. Ethylene oxide linked oligonucleosides are prepared as described in U.S. Pat. No. 5,223,618, herein incorporated by reference.

RNA Synthesis

In general, RNA synthesis chemistry is based on the selective incorporation of various protecting groups at strategic intermediary reactions. Although one of ordinary skill in the art will understand the use of protecting groups in organic synthesis, a useful class of protecting groups includes silyl ethers. In particular bulky silyl ethers are used to protect the 5′-hydroxyl in combination with an acid-labile orthoester protecting group on the 2′hydroxyl. This set of protecting groups is then used with standard solid-phase synthesis technology. It is important to lastly remove the acid labile orthoester protecting group after all other synthetic steps. Moreover, the early use of the silyl protecting groups during synthesis ensures facile removal when desired, without undesired deprotection of 2′ hydroxyl. Following this procedure for the sequential protection of the 5′-hydroxyl in combination with protection of the 2′-hydroxyl by protecting groups that are differentially removed and are differentially chemically labile, RNA oligonucleotides were synthesized. RNA oligonucleotides are synthesized in a stepwise fashion. Each nucleotide is added sequentially (3′- to 5′-direction) to a solid support-bound oligonucleotide. The first nucleoside at the 3′-end of the chain is covalently attached to a solid support. The nucleotide precursor, a ribonucleoside phosphoramidite, and activator are added, coupling the second base onto the 5′-end of the first nucleoside. The support is washed and any unreacted 5′-hydroxyl groups are capped with acetic anhydride to yield 5′-acetyl moieties. The linkage is then oxidized to the more stable and ultimately desired P(V) linkage. At the end of the nucleotide addition cycle, the 5′-silyl group is cleaved with fluoride. The cycle is repeated for each subsequent nucleotide.

Following synthesis, the methyl protecting groups on the phosphates are cleaved in 30 minutes utilizing 1 M disodium-2-carbamoyl-2-cyanoethylene-1,1-dithiolate trihydrate (S2Na2) in DMF. The deprotection solution is washed from the solid supportbound oligonucleotide using water. The support is then treated with 40% methylamine in water for 10 minutes at 55 ° C. This releases the RNA oligonucleotides into solution, deprotects the exocyclic amines, and modifies the 2′-groups. The oligonucleotides can be analyzed by anion exchange HPLC at this stage.

The 2′-orthoester groups are the last protecting groups to be removed. The ethylene glycol monoacetate orthoester protecting group developed by Dharmacon Research, Inc. (Lafayette, Colo.), is one example of a useful orthoester protecting group which, has the following important properties. It is stable to the conditions of nucleoside phosphoramidite synthesis and oligonucleotide synthesis. However, after oligonucleotide synthesis the oligonucleotide is treated with methylamine which not only cleaves the oligonucleotide from the solid support but also removes the acetyl groups from the orthoesters. The resulting 2-ethyl-hydroxyl substituents on the orthoester are less electron withdrawing than the acetylated precursor. As a result, the modified orthoester becomes more labile to acid-catalyzed hydrolysis. Specifically, the rate of cleavage is approximately 10 times faster after the acetyl groups are removed, Therefore, this orthoester possesses sufficient stability in order to be compatible with oligonucleotide synthesis and yet, when subsequently modified, permits deprotection to be carried out under relatively mild aqueous conditions compatible with the final RNA oligonucleotide product. Additionally, methods of RNA synthesis are well known in the art (Scaringe, S. A. Ph.D. Thesis, University of Colorado, 1996; Scaringe, S. A., et al., J. Am. Chem. Soc., 1″8, 120, 11820-11821; Matteucci, M. D. and Caruthers, M. H. J. Am. Chem. Soc., 1981, 103, 3185-3191; Beaucage, S. L. and Caruthers, M. H. Tetrahedron Lett., 1981, 22, 1859-1862; Dahl, B. J., et al., Acta Chem. Scand,. 1990, 44, 639-641; Reddy, M. P., et al., Tetrahedrom Lett., 1994, 25, 4311-4314; Wincott, F. et al., Nucleic Acids Res., 1995, 23, 2677-2684; Griffin, B. E., et al., Tetrahedron, 1967, 23, 2301-2313; Griffin, B. E., et al., Tetrahedron, 1967, 23, 2315-2331).

RNA antisense compounds (RNA oligonucleotides) of the present invention can be synthesized by the methods herein or purchased from Dharmacon Research, Inc (Lafayette, Colo.). Once synthesized, complementary RNA antisense compounds can then be annealed by methods known in the art to form double stranded (duplexed) antisense compounds. For example, duplexes can be formed by combining 30 μl of each of the complementary strands of RNA oligonucleotides (50 uM RNA oligonucleotide solution) and 15 tl of 5× annealing buffer (100 mM potassium acetate, 30 mM HEPES-KOH pH 7.4, 2 mM magnesium acetate) followed by heating for 1 minute at 90° C., then 1 hour at 37° C. The resulting duplexed antisense compounds can be used in kits, assays, screens, or other methods to investigate the role of a target nucleic acid.

Synthesis of Chimeric Oligonucleotides

Chimeric oligonucleotides, oligonucleosides or mixed oligonucleotides/oligonucleosides of the invention can be of several different types. These include a first type wherein the “gap” segment of linked nucleosides is positioned between 5′ and 3′ “wing” segments of linked nucleosides and a second “open end” type wherein the “gap” segment is located at either the 3′ or the 5′ terminus of the oligomeric compound. Oligonucleotides of the first type are also known in the art as “gapmers” or gapped oligonucleotides. Oligonucleotides of the second type are also known in the art as “hemimers” or “wingmers”.

[2′-O-Me]-[2′-deoxy]-[2′-O-Me]Chimeric Phosphorothioate Oligonucleotides

Chimeric oligonucleotides having 2′-O-alkyl phosphorothioate and 2′-deoxy phosphorothioate oligonucleotide segments are synthesized using an Applied Biosystems automated DNA synthesizer Model 394, as above. Oligonucleotides are synthesized using the automated synthesizer and 2′-deoxy-5′-dimethoxytrityl-3′-O-phosphoramidite for the DNA portion and 5′-dimethoxytrityl-2′-O-methyl-3′-O-phosphoramidite for 5′ and 3′ wings. The standard synthesis cycle is modified by incorporating coupling steps with increased reaction times for the 5′-dimethoxytrityl-2′-O-methyl-3′-O-phosphoramidite. The fully protected oligonucleotide is cleaved from the support and deprotected in concentrated ammonia (NH4OH) for 12-16 hr at 55° C. The deprotected oligo is then recovered by an appropriate method (precipitation, column chromatography, volume reduced in vacuo and analyzed spetrophotometrically for yield and for purity by capillary electrophoresis and by mass spectrometry.

[2′-O-(2-Methoxyethyl)]-[2′-deoxy]; -[2′-O-(Methoxyethyl)]Chimeric Phosphorothioate Oligonucleotides

[2′-O-(2-methoxyethyl)]-[2′-deoxy]-[-2′-O-(methoxyethyl)] chimeric phosphorothioate oligonucleotides were prepared as per the procedure above for the 2′0-methyl chimeric oligonucleotide, with the substitution of 2′-O-(methoxyethyl)amidites for the 2′-O-methyl amidites.

[2′-O-(2-Methoxyethyl)Phosphodiester]-[2′-deoxy Phosphorothioate]-[2′-O-(2-Methoxyethyl)Phosphodiester]Chimeric Oligonucleotides

[2′-O-(2-methoxyethyl phosphodiester]-[2′-deoxy phosphorothioate]-[2′-O(methoxyethyl)phosphodiester]chimeric oligonucleotides are prepared as per the above procedure for the 2′-O-methyl chimeric oligonucleotide with the substitution of 2′-O(methoxyethyl)amidites for the 2′-O-methyl amidites, oxidation with iodine to generate the phosphodiester internucleotide linkages within the wing portions of the chimeric structures and sulfurization utilizing 3,H-1,2 benzodithiole-3-one 1,1 dioxide (Beaucage Reagent) to generate the phosphorothioate internucleotide linkages for the center gap.

Other chimeric oligonucleotides, chimeric oligonucleosides and mixed chimeric oligonucleotides/oligonucleosides are synthesized according to U.S. Pat. No. 5,623,065, herein incorporated by reference.

The methods of the present invention are particularly useful in antisense therapeutics. It is not necessary that the antisense target be associated with liver disease per se, since many current antisense targets are expressed to high levels in liver and other organs. In particular, targets associated with metabolic and cardiovascular diseases and conditions are particularly amenable to knockdown in the liver and have been shown in animals and in clinical studies to have therapeutic effects).

The compounds of the invention may also be admixed, encapsulated, conjugated or otherwise associated with other molecules, molecule structures or mixtures of compounds, as for example, liposomes, receptor-targeted molecules, oral, rectal, topical or other formulations, for assisting in uptake, distribution and/or absorption. The antisense compounds of the invention encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal, including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to prodrugs and pharmaceutically acceptable salts of the compounds of the invention, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.

Claims

1. A method of reducing expression of a target RNA in an animal, in need of reducing expression of said target RNA, comprising administering to said animal a gap-widened antisense oligonucleotide 18-24 linked nucleosides in length comprising: wherein the gap region is located between said first wing region and said second wing region and, wherein each nucleoside of said first and second wing region is a 2′modified nucleoside thereby reducing expression of said target RNA in said animal.

(a) a gap region having 12 to 18contiguous 2′-deoxyribonucleosides; and
(b) a first wing region having 1 to 4 contiguous nucleosides; and
(c) a second wing region having 1 to 4 contiguous nucleosides;

2.-3. (canceled)

4. The method of claim 1, wherein the target RNA is associated with a metabolic or a cardiovascular disease or condition.

5. The method of claim 1, wherein the metabolic disease or condition is selected from metabolic syndrome, diabetes, obesity, hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, Type 2 diabetes, diet-induced obesity, hyperglycemia, insulin resistance, hepatic steatosis, fatty liver disease, or non-alcoholic steatohepatitis.

6. The method of claim 1, wherein the cardiovascular disease or condition is selected from familial hypercholesterolemia, nonfamilial hypercholesterolemia, mixed dyslipidemia, dysbetalipoproteinemia, atherosclerosis, coronary artery disease, myocardial infarction, hypertension, carotid artery diseases, carotid artery disease, stroke, cerebrovascular disease, peripheral vascular disease, thrombosis, or arterial aneurism.

7. The method of claim 1, wherein the gap-widened antisense oligonucleotide has a wing-gap-wing motif selected from 2-16-2, 3-14-3, 2-14-2, 3-12-3 or 4-12-4.

8. The method of claim 1, wherein the gap-widened antisense oligonucleotide has at least one phosphorothioate internucleotide linkage.

9. The method of claim 6, wherein the gap-widened antisense oligonucleotide has all phosphorothioate internucleotide linkages.

10. The method of claim 1, wherein gap-widened antisense oligonucleotide has at least one 5-methylcytosine.

11.-25. (canceled)

26. A method of modulating gene expression in an animal comprising the step of contacting said animal with the pharmaceutical composition comprising a gap-widened antisense oligoncuelotide 18-24 linked nucleosides in length comprising: wherein the gap region is located between said first wing region and said second wing region and, wherein each nucleoside of said first and second wing region is a 2′ modified nucleoside thereby modulating gene expression in said animal.

(a) a gap region having 12 to 18 contiguous 2′-deoxyribonucleotides;
(b) a first wing region having 1 to 4 contiguous nucleosides; and
(c) a second wing region having 1 to 4 contiguous nucleosides;

27.-28. (canceled)

Patent History
Publication number: 20110207797
Type: Application
Filed: Jan 18, 2011
Publication Date: Aug 25, 2011
Applicant: Isis Pharmaceuticals, Inc. (Carlsbad, CA)
Inventors: Brett P. Monia (Encinitas, CA), Andrew M. Siwkowski (Encinitas, CA), Sanjay Bhanot (Carlsbad, CA)
Application Number: 13/008,704
Classifications
Current U.S. Class: 514/44.0A
International Classification: A61K 31/7088 (20060101); A61P 9/00 (20060101); A61P 3/00 (20060101); A61P 3/10 (20060101); A61P 3/04 (20060101); A61P 3/06 (20060101); A61P 1/16 (20060101); A61P 9/10 (20060101); A61P 9/12 (20060101); A61P 7/02 (20060101);